Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biomed Pharmacother ; 174: 116526, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38574621

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a debilitating neurodegenerative disorder of the cerebellum and brainstem. Memantine has been proposed as a potential treatment for SCA1. It blocks N-methyl-D-aspartate (NMDA) receptors on neurons, reduces excitotoxicity and decreases neurodegeneration in Alzheimer models. However, in cerebellar neurodegenerative diseases, the potential value of memantine is still unclear. We investigated the effects of memantine on motor performance and synaptic transmission in the cerebellum in a mouse model where mutant ataxin 1 is specifically targeted to glia. Lentiviral vectors (LVV) were used to express mutant ataxin 1 selectively in Bergmann glia (BG). In mice transduced with the mutant ataxin 1, chronic treatment with memantine improved motor activity during initial tests, presumably due to preserved BG and Purkinje cell (PC) morphology and numbers. However, mice were unable to improve their rota rod scores during next days of training. Memantine also compromised improvement in the rota rod scores in control mice upon repetitive training. These effects may be due to the effects of memantine on plasticity (LTD suppression) and NMDA receptor modulation. Some effects of chronically administered memantine persisted even after its wash-out from brain slices. Chronic memantine reduced morphological signs of neurodegeneration in the cerebellum of SCA1 model mice. This resulted in an apparent initial reduction of ataxic phenotype, but memantine also affected cerebellar plasticity and ultimately compromised motor learning. We speculate that that clinical application of memantine in SCA1 might be hampered by its ability to suppress NMDA-dependent plasticity in cerebellar cortex.


Assuntos
Modelos Animais de Doenças , Memantina , Fenótipo , Ataxias Espinocerebelares , Animais , Memantina/farmacologia , Ataxias Espinocerebelares/tratamento farmacológico , Ataxias Espinocerebelares/patologia , Camundongos , Ataxina-1/metabolismo , Ataxina-1/genética , Atividade Motora/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Cerebelo/patologia , Cerebelo/metabolismo , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/patologia , Células de Purkinje/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Camundongos Transgênicos , Camundongos Endogâmicos C57BL , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Neuroglia/metabolismo , Masculino , Plasticidade Neuronal/efeitos dos fármacos
2.
Cells ; 11(14)2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35883691

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is an intractable progressive neurodegenerative disease that leads to a range of movement and motor defects and is eventually lethal. Purkinje cells (PC) are typically the first to show signs of degeneration. SCA1 is caused by an expansion of the polyglutamine tract in the ATXN1 gene and the subsequent buildup of mutant Ataxin-1 protein. In addition to its toxicity, mutant Ataxin-1 protein interferes with gene expression and signal transduction in cells. Recently, it is evident that ATXN1 is not only expressed in neurons but also in glia, however, it is unclear the extent to which either contributes to the overall pathology of SCA1. There are various ways to model SCA1 in mice. Here, functional deficits at cerebellar synapses were investigated in two mouse models of SCA1 in which mutant ATXN1 is either nonspecifically expressed in all cell types of the cerebellum (SCA1 knock-in (KI)), or specifically in Bergmann glia with lentiviral vectors expressing mutant ATXN1 under the control of the astrocyte-specific GFAP promoter. We report impairment of motor performance in both SCA1 models. In both cases, prominent signs of astrocytosis were found using immunohistochemistry. Electrophysiological experiments revealed alteration of presynaptic plasticity at synapses between parallel fibers and PCs, and climbing fibers and PCs in SCA1 KI mice, which is not observed in animals expressing mutant ATXN1 solely in Bergmann glia. In contrast, short- and long-term synaptic plasticity was affected in both SCA1 KI mice and glia-targeted SCA1 mice. Thus, non-neuronal mechanisms may underlie some aspects of SCA1 pathology in the cerebellum. By combining the outcomes of our current work with our previous data from the B05 SCA1 model, we further our understanding of the mechanisms of SCA1.


Assuntos
Ataxias Espinocerebelares , Animais , Ataxina-1/genética , Ataxina-1/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal , Células de Purkinje , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia
3.
Brain Sci ; 12(4)2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35448027

RESUMO

Memantine is an FDA approved drug for the treatment of Alzheimer's disease. It reduces neurodegeneration in the hippocampus and cerebral cortex through the inhibition of extrasynaptic NMDA receptors in patients and mouse models. Potentially, it could prevent neurodegeneration in other brain areas and caused by other diseases. We previously used memantine to prevent functional damage and to retain morphology of cerebellar neurons and Bergmann glia in an optogenetic mouse model of spinocerebellar ataxia type-1 (SCA1). However, before suggesting wider use of memantine in clinics, its side effects must be carefully evaluated. Blockers of NMDA receptors are controversial in terms of their effects on anxiety. Here, we investigated the effects of chronic application of memantine over 9 weeks to CD1 mice and examined rotarod performance and anxiety-related behaviors. Memantine-treated mice exhibited an inability to adapt to anxiety-causing conditions which strongly affected their rotarod performance. A tail suspension test revealed increased signs of behavioral despair. These data provide further insights into the potential deleterious effects of memantine which may result from the lack of adaptation to novel, stressful conditions. This effect of memantine may affect the results of tests used to assess motor performance and should be considered during clinical trials of memantine in patients.

4.
Food Chem ; 373(Pt B): 131507, 2022 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-34763933

RESUMO

Present study investigated effect of dietary buckwheat in alleviating bisphenol A (BPA) mediated oxidative stress, concomitant sirtuin1 levels in serum, stomach, and liver of rats. Experimental group A and B ingested standard diet, C and D consumed buckwheat (30%); group A and C drank normal water, B and C had BPA contamination (10 mg L-1). Sirtuin1 mean B/A ratio nearing unity in all tissues reveals inertness of BPA towards sirtuin1. Dietary buckwheat improved sirtuin1 levels both in normal (mean C/A ratio of serum, 1.65; liver, 1.24; stomach, 1.78) and BPA fed state (mean D/B ratio of serum, 1.9; liver, 1.26; stomach, 1.75). Buckwheat augmented antioxidant status in BPA fed rats as seen in mean D/B ratio of serum (catalase, 2.4; glutathione reductase (GR), 1.33; Thiols, 1.2), liver (catalase, 2; GR, 2.5; Thiols, 1.36) and stomach (catalase, 1.31; GR, 1.5; Thiols, 1.33). Therefore, buckwheat counters BPA-led oxidative stress and modulates sirtuin1.


Assuntos
Antioxidantes , Fagopyrum , Animais , Antioxidantes/metabolismo , Compostos Benzidrílicos/metabolismo , Catalase/metabolismo , Dieta , Fagopyrum/metabolismo , Fígado/metabolismo , Estresse Oxidativo , Fenóis , Ratos , Sirtuína 1/genética , Sirtuína 1/metabolismo
5.
Int J Mol Sci ; 22(21)2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34769018

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a devastating neurodegenerative disorder. In recent years, attention of researchers has increasingly been focused on studying the role of brain insulin resistance (BIR) in the AD pathogenesis. Neuroinflammation makes a significant contribution to the BIR due to the activation of NLRP3 inflammasome. This study was devoted to the understanding of the potential therapeutic roles of the NLRP3 inflammasome in neurodegeneration occurring concomitant with BIR and its contribution to the progression of emotional disorders. METHODS: To test the impact of innate immune signaling on the changes induced by Aß1-42 injection, we analyzed animals carrying a genetic deletion of the Nlrp3 gene. Thus, we studied the role of NLRP3 inflammasomes in health and neurodegeneration in maintaining brain insulin signaling using behavioral, electrophysiological approaches, immunohistochemistry, ELISA and real-time PCR. RESULTS: We revealed that NLRP3 inflammasomes are required for insulin-dependent glucose transport in the brain and memory consolidation. Conclusions NLRP3 knockout protects mice against the development of BIR: Taken together, our data reveal the protective role of Nlrp3 deletion in the regulation of fear memory and the development of Aß-induced insulin resistance, providing a novel target for the clinical treatment of this disorder.


Assuntos
Doença de Alzheimer/metabolismo , Inflamassomos/metabolismo , Resistência à Insulina/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Neuroinflamatórias/metabolismo
6.
Int J Mol Sci ; 22(15)2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34360588

RESUMO

Spinocerebellar ataxias are a family of fatal inherited diseases affecting the brain. Although specific mutated proteins are different, they may have a common pathogenetic mechanism, such as insufficient glutamate clearance. This function fails in reactive glia, leading to excitotoxicity and overactivation of NMDA receptors. Therefore, NMDA receptor blockers could be considered for the management of excitotoxicity. One such drug, memantine, currently used for the treatment of Alzheimer's disease, could potentially be used for the treatment of other forms of neurodegeneration, for example, spinocerebellar ataxias (SCA). We previously demonstrated close parallels between optogenetically induced cerebellar degeneration and SCA1. Here we induced reactive transformation of cerebellar Bergmann glia (BG) using this novel optogenetic approach and tested whether memantine could counteract changes in BG and Purkinje cell (PC) morphology and expression of the main glial glutamate transporter-excitatory amino acid transporter 1 (EAAT1). Reactive BG induced by chronic optogenetic stimulation presented increased GFAP immunoreactivity, increased thickness and decreased length of its processes. Oral memantine (~90 mg/kg/day for 4 days) prevented thickening of the processes (1.57 to 1.81 vs. 1.62 µm) and strongly antagonized light-induced reduction in their average length (186.0 to 150.8 vs. 171.9 µm). Memantine also prevented the loss of the key glial glutamate transporter EAAT1 on BG. Finally, memantine reduced the loss of PC (4.2 ± 0.2 to 3.2 ± 0.2 vs. 4.1 ± 0.3 cells per 100 µm of the PC layer). These results identify memantine as potential neuroprotective therapeutics for cerebellar ataxias.


Assuntos
Dopaminérgicos/farmacologia , Memantina/farmacologia , Doenças Neurodegenerativas/prevenção & controle , Neuroglia/efeitos dos fármacos , Optogenética/efeitos adversos , Substâncias Protetoras/farmacologia , Células de Purkinje/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Camundongos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/patologia , Neuroglia/patologia , Células de Purkinje/patologia
7.
Biochemistry (Mosc) ; 86(6): 746-760, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34225598

RESUMO

Blood-brain barrier (BBB) is a structural and functional element of the neurovascular unit (NVU), which includes cells of neuronal, glial, and endothelial nature. The main functions of NVU include maintenance of the control of metabolism and chemical homeostasis in the brain tissue, ensuring adequate blood flow in active regions, regulation of neuroplasticity processes, which is realized through intercellular interactions under normal conditions, under stress, in neurodegeneration, neuroinfection, and neurodevelopmental diseases. Current versions of the BBB and NVU models, static and dynamic, have significantly expanded research capabilities, but a number of issues remain unresolved, in particular, personification of the models for a patient. In addition, application of both static and dynamic models has an important problem associated with the difficulty in reproducing pathophysiological mechanisms responsible for the damage of the structural and functional integrity of the barrier in the diseases of the central nervous system. More knowledge on the cellular and molecular mechanisms of BBB and NVU damage in pathology is required to solve this problem. This review discusses current state of the cellular and molecular mechanisms that control BBB permeability, pathobiochemical mechanisms and manifestations of BBB breakdown in stress and neurodegenerative diseases, as well as the problems and prospects of creating in vitro BBB and NVU models for translational studies in neurology and neuropharmacology. Deciphering BBB (patho)physiology will open up new opportunities for further development in the related areas of medicine such as regenerative medicine, neuropharmacology, and neurorehabilitation.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Doenças Neurodegenerativas/fisiopatologia , Estresse Psicológico/fisiopatologia , Barreira Hematoencefálica/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo , Estresse Psicológico/metabolismo
8.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-33925080

RESUMO

Pathophysiology of chronic neurodegeneration is mainly based on complex mechanisms related to aberrant signal transduction, excitation/inhibition imbalance, excitotoxicity, synaptic dysfunction, oxidative stress, proteotoxicity and protein misfolding, local insulin resistance and metabolic dysfunction, excessive cell death, development of glia-supported neuroinflammation, and failure of neurogenesis. These mechanisms tightly associate with dramatic alterations in the structure and activity of the neurovascular unit (NVU) and the blood-brain barrier (BBB). NVU is an ensemble of brain cells (brain microvessel endothelial cells (BMECs), astrocytes, pericytes, neurons, and microglia) serving for the adjustment of cell-to-cell interactions, metabolic coupling, local microcirculation, and neuronal excitability to the actual needs of the brain. The part of the NVU known as a BBB controls selective access of endogenous and exogenous molecules to the brain tissue and efflux of metabolites to the blood, thereby providing maintenance of brain chemical homeostasis critical for efficient signal transduction and brain plasticity. In Alzheimer's disease, mitochondria are the target organelles for amyloid-induced neurodegeneration and alterations in NVU metabolic coupling or BBB breakdown. In this review we discuss understandings on mitochondria-driven NVU and BBB dysfunction, and how it might be studied in current and prospective NVU/BBB in vitro models for finding new approaches for the efficient pharmacotherapy of Alzheimer's disease.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Mitocôndrias/fisiologia , Modelos Neurológicos , Degeneração Neural/etiologia , Degeneração Neural/fisiopatologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/fisiopatologia , Animais , Dano ao DNA , DNA Mitocondrial/metabolismo , Humanos , Técnicas In Vitro , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/fisiologia , Espécies Reativas de Oxigênio/metabolismo
9.
Neurobiol Dis ; 154: 105340, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33753288

RESUMO

Bergmann glia (BG) are highly specialized radial astrocytes of the cerebellar cortex, which play a key role in the uptake of synaptic glutamate via the excitatory amino acid transporter EAAT1. Multiple lines of evidence suggest that in cerebellar neurodegenerative diseases reactive BG has a negative impact on neuronal function and survival through compromised EAAT activity. A family of such diseases are those caused by expansion of CAG repeats in genes of the ataxin family, resulting in spinocerebellar ataxias (SCA). We investigated the contribution of BG to the pathogenesis of cerebellar neurodegeneration in a model of SCA1, which was induced by expression of a polyglutamine mutant of ataxin-1 (ATXN1[Q85]) in BG specifically. We compared the outcomes with a novel model where we triggered excitotoxicity by a chronic optogenetic activation of BG with channelrhodopsin-2 (ChR2). In both cases we detected evidence of reduced glutamate uptake manifested by prolongation of excitatory postsynaptic currents in Purkinje cells which is consistent with documented reduction of expression and/or function of EAAT1. In both models we detected astroglyosis and Purkinje cells atrophy. Finally, the same pattern was detected in a knock-in mouse which expresses a polyglutamine mutant ataxin-1 ATXN1[Q154] in a non-cell-selective manner. Our results suggest that ATXN1[Q85] and ChR2-induced insult targeted to BG closely mimics SCA1 pathology, where excessive glutamate signaling appears to be a common feature likely being an important contributor to cerebellar neurodegeneration.


Assuntos
Ataxina-1/biossíntese , Transportador 1 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 1 de Aminoácido Excitatório/biossíntese , Neuroglia/metabolismo , Optogenética/efeitos adversos , Células de Purkinje/metabolismo , Animais , Ataxina-1/genética , Morte Celular/fisiologia , Transportador 1 de Aminoácido Excitatório/genética , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/patologia , Estimulação Luminosa/efeitos adversos , Células de Purkinje/patologia
10.
Brain Res ; 1752: 147220, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33358726

RESUMO

Neuroinflammation has been classified as a trigger of behavioral alterations and cognitive impairments in many neurological conditions, including Alzheimer's disease, major depression, anxiety and others. Regardless of the cause of neuroinflammation, key molecules, which sense neuropathological conditions, are intracellular multiprotein signaling inflammasomes. Increasing evidence shows that the inflammatory response, mediated by activated nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasomes, is associated with the onset and progression of a wide range of diseases of the CNS. However, whether the NLRP3 inflammasome in the CNS is involved in the learning, development of anxiety and adult neurogenesis remains elusive. Therefore, the present study was designed to assess NLRP3 inflammasome contribution in anxiety and reveal its potential involvement in the experimental acquisition of fear responses and hippocampal neurogenesis. Behavioral, immunohistochemical and electrophysiological alterations were measured to evaluate role of neuroinflammation in the limbic system of mice. In this study, we describe interrelated neurophysiological mechanisms, which culminate in absence of NLRP3 inflammasome in young 4 months mice. These include the following: anxious behavior and deterioration in learning and memory of fear conditioning; impairment of adult neurogenesis; reduction and altered morphology of astrocytes in the brain; hyperexcitability in basolateral amygdala (BLA); impaired activation in axons of pyramidal cells of CA1 hippocampal zone in NLRP3 KO mice particularly via the Schaffer collateral pathway; and impaired synaptic transduction in pyramidal cells mediated by an embarrassment of neurotransmitter release from presynaptic site in CA3 hippocampal zone. The present study has demonstrated the novel findings that basal level of NLRP3 inflammasome in the brain of young mice is required for conditioning-induced plasticity in the ventral hippocampus and the basolateral amygdala. The deletion of NLRP3 impair synaptic transduction and caused anxiety-like behavior and labored fear learning, suggesting that low grade inflammation, mediated by NLRP3 expression, play a key role in memory consolidation.


Assuntos
Ansiedade/fisiopatologia , Encefalite/fisiopatologia , Hipocampo/fisiopatologia , Inflamassomos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Animais , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo
11.
Rev Neurosci ; 30(8): 807-820, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31152644

RESUMO

The excitation/inhibition (E/I) balance controls the synaptic inputs to prevent the inappropriate responses of neurons to input strength, and is required to restore the initial pattern of network activity. Various neurotransmitters affect synaptic plasticity within neural networks via the modulation of neuronal E/I balance in the developing and adult brain. Less is known about the role of E/I balance in the control of the development of the neural stem and progenitor cells in the course of neurogenesis and gliogenesis. Recent findings suggest that neural stem and progenitor cells appear to be the target for the action of GABA within the neurogenic or oligovascular niches. The same might be true for the role of neuropeptides (i.e. oxytocin) in neurogenic niches. This review covers current understanding of the role of E/I balance in the regulation of neuroplasticity associated with social behavior in normal brain, and in neurodevelopmental and neurodegenerative diseases. Further studies are required to decipher the GABA-mediated regulation of postnatal neurogenesis and synaptic integration of newly-born neurons as a potential target for the treatment of brain diseases.


Assuntos
Doenças Neurodegenerativas/etiologia , Transtornos do Neurodesenvolvimento/etiologia , Neurogênese , Potenciais Sinápticos , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/fisiologia , Humanos , Doenças Neurodegenerativas/fisiopatologia , Transtornos do Neurodesenvolvimento/fisiopatologia
12.
Brain Sci ; 9(4)2019 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-31013844

RESUMO

Astrogliosis is a pathological process that affects the density, morphology, and function of astrocytes. It is a common feature of brain trauma, autoimmune diseases, and neurodegeneration including spinocerebellar ataxia type 1 (SCA1), a poorly understood neurodegenerative disease. S100ß is a Ca2+ binding protein. In SCA1, excessive excretion of S100ß by reactive astrocytes and its uptake by Purkinje cells has been demonstrated previously. Under pathological conditions, excessive extracellular concentration of S100ß stimulates the production of proinflammatory cytokines and induces apoptosis. We modeled astrogliosis by S100ß injections into cerebellar cortex in mice. Injections of S100ß led to significant changes in Bergmann glia (BG) cortical organization and affected their processes. S100ß also changed morphology of the Purkinje cells (PCs), causing a significant reduction in the dendritic length. Moreover, the short-term synaptic plasticity and depolarization-induced suppression of synaptic transmission were disrupted after S100ß injections. We speculate that these effects are the result of Ca2+-chelating properties of S100ß protein. In summary, exogenous S100ß induced astrogliosis in cerebellum could lead to neuronal dysfunction, which resembles a natural neurodegenerative process. We suggest that astrocytes play an essential role in SCA1 pathology, and that astrocytic S100ß is an important contributor to this process.

13.
Front Physiol ; 9: 1656, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30534080

RESUMO

Adipose tissue is recognized as an important organ with metabolic, regulatory, and plastic roles. Adipose tissue-derived stem cells (ASCs) with self-renewal properties localize in the stromal vascular fraction (SVF) being present in a vascular niche, thereby, contributing to local regulation of angiogenesis and vessel remodeling. In the past decades, ASCs have attracted much attention from biologists and bioengineers, particularly, because of their multilineage differentiation potential, strong proliferation, and migration abilities in vitro and high resistance to oxidative stress and senescence. Current data suggest that the SVF serves as an important source of endothelial progenitors, endothelial cells, and pericytes, thereby, contributing to vessel remodeling and growth. In addition, ASCs demonstrate intriguing metabolic and interlineage plasticity, which makes them good candidates for creating regenerative therapeutic protocols, in vitro tissue models and microphysiological systems, and tissue-on-chip devices for diagnostic and regeneration-supporting purposes. This review covers recent achievements in understanding the metabolic activity within the SVF niches (lactate and NAD+ metabolism), which is critical for maintaining the pool of ASCs, and discloses their pro-angiogenic potential, particularly, in the complex therapy of cardiovascular and cerebrovascular diseases.

14.
J Neurochem ; 143(6): 660-670, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29049849

RESUMO

Cerebellar Purkinje cells (PCs) express two members of the classical protein kinase C (cPKC) subfamily, namely, PKCα and PKCγ. Previous studies on PKCγ knockout (KO) mice have revealed a critical role of PKCγ in the pruning of climbing fibers (CFs) from PCs during development. The question remains as to why only PKCγ and not PKCα is involved in CF synapse elimination from PCs. To address this question, we assessed the expression levels of PKCγ and PKCα in wild-type (WT) and PKCγ KO PCs using PC-specific quantitative real-time reverse transcription-polymerase chain reaction, western blotting, and immunohistochemical analysis. The results revealed that the vast majority of cPKCs in PCs were PKCγ, whereas PKCα accounted for the remaining minimal fraction. The amount of PKCα was not up-regulated in PKCγ KO PCs. Lentiviral expression of PKCα in PKCγ KO PCs resulted in a 10-times increase in the amount of PKCα mRNA in the PKCγ KO PCs, compared to that in WT PCs. Our quantification showed that the expression levels of cPKC mRNA in PKCγ KO PCs increased roughly from 1% to 22% of that in WT PCs solely through PKCα expression. The up-regulation of PKCα in PKCγ KO PCs significantly rescued the impaired CF synapse elimination. Although both PKCα and PKCγ are capable of pruning supernumerary CF synapses from developing PCs, these results suggest that the expression levels of cPKCs in PKCγ KO PCs are too low for CF pruning.


Assuntos
Cerebelo/enzimologia , Cerebelo/crescimento & desenvolvimento , Proteína Quinase C/biossíntese , Células de Purkinje/citologia , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Nervosas/metabolismo , Isoformas de Proteínas , Transcriptoma
15.
J Physiol ; 595(1): 141-164, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440721

RESUMO

KEY POINTS: Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disease caused by a gene defect, leading to movement disorder such as cerebellar ataxia. It remains largely unknown which functional defect contributes to the cerebellar ataxic phenotype in SCA1. In this study, we report progressive dysfunction of metabotropic glutamate receptor (mGluR) signalling, which leads to smaller slow synaptic responses, reduced dendritic Ca2+ signals and impaired synaptic plasticity at cerebellar synapses, in the early disease stage of SCA1 model mice. We also show that enhancement of mGluR signalling by a clinically available drug, baclofen, leads to improvement of motor performance in SCA1 mice. SCA1 is an incurable disease with no effective treatment, and our results may provide mechanistic grounds for targeting mGluRs and a novel drug therapy with baclofen to treat SCA1 patients in the future. ABSTRACT: Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disease that presents with cerebellar ataxia and motor learning defects. Previous studies have indicated that the pathology of SCA1, as well as other ataxic diseases, is related to signalling pathways mediated by the metabotropic glutamate receptor type 1 (mGluR1), which is indispensable for proper motor coordination and learning. However, the functional contribution of mGluR signalling to SCA1 pathology is unclear. In the present study, we show that SCA1 model mice develop a functional impairment of mGluR signalling which mediates slow synaptic responses, dendritic Ca2+ signals, and short- and long-term synaptic plasticity at parallel fibre (PF)-Purkinje cell (PC) synapses in a progressive manner from the early disease stage (5 postnatal weeks) prior to PC death. Notably, impairment of mGluR-mediated dendritic Ca2+ signals linearly correlated with a reduction of PC capacitance (cell surface area) in disease progression. Enhancement of mGluR signalling by baclofen, a clinically available GABAB receptor agonist, led to an improvement of motor performance in SCA1 mice and the improvement lasted ∼1 week after a single application of baclofen. Moreover, the restoration of motor performance in baclofen-treated SCA1 mice matched the functional recovery of mGluR-mediated slow synaptic currents and mGluR-dependent short- and long-term synaptic plasticity. These results suggest that impairment of synaptic mGluR cascades is one of the important contributing factors to cerebellar ataxia in early and middle stages of SCA1 pathology, and that modulation of mGluR signalling by baclofen or other clinical interventions may be therapeutic targets to treat SCA1.


Assuntos
Cerebelo/fisiopatologia , Receptores de Glutamato Metabotrópico/fisiologia , Ataxias Espinocerebelares/fisiopatologia , Animais , Baclofeno/farmacologia , Baclofeno/uso terapêutico , Fenômenos Biomecânicos , Cálcio/fisiologia , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores , Feminino , Agonistas dos Receptores de GABA-B/farmacologia , Agonistas dos Receptores de GABA-B/uso terapêutico , Membro Posterior/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Teste de Desempenho do Rota-Rod , Transdução de Sinais , Ataxias Espinocerebelares/tratamento farmacológico
16.
Front Physiol ; 7: 599, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27990124

RESUMO

Currently, there is a considerable interest to the assessment of blood-brain barrier (BBB) development as a part of cerebral angiogenesis developmental program. Embryonic and adult angiogenesis in the brain is governed by the coordinated activity of endothelial progenitor cells, brain microvascular endothelial cells, and non-endothelial cells contributing to the establishment of the BBB (pericytes, astrocytes, neurons). Metabolic and functional plasticity of endothelial progenitor cells controls their timely recruitment, precise homing to the brain microvessels, and efficient support of brain angiogenesis. Deciphering endothelial progenitor cells physiology would provide novel engineering approaches to establish adequate microfluidically-supported BBB models and brain microphysiological systems for translational studies.

17.
Rev Neurosci ; 27(4): 365-76, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26641963

RESUMO

Patch clamp is a golden standard for studying (patho)physiological processes affecting membranes of excitable cells. This method is rather labor-intensive and requires well-trained professionals and long-lasting experimental procedures; therefore, accurate designing of the experiments with patch clamp methodology as well as collecting and analyzing the data obtained are essential for the widely spread implementation of this method into the routine research practice. Recently, the method became very prospective not only for the characterization of single excitable cells but also for the detailed assessment of intercellular communication, i.e. within the neurovascular unit. Here, we analyze the main advantages and disadvantages of patch clamp method, with special focus on the tendencies in clamping technique improvement with the help of patch electrodes for the assessment of intercellular communication in the brain.


Assuntos
Encéfalo/fisiologia , Comunicação Celular/fisiologia , Eletrofisiologia , Acoplamento Neurovascular/fisiologia , Potenciais de Ação/fisiologia , Animais , Eletrofisiologia/métodos , Humanos , Técnicas de Patch-Clamp/métodos
18.
Hum Mol Genet ; 24(2): 525-39, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25217572

RESUMO

Amyloid assemblies are associated with a wide range of human disorders, including Alzheimer's and Parkinson's diseases. Here, we identify protein kinase C (PKC) γ, a serine/threonine kinase mutated in the neurodegenerative disease spinocerebellar ataxia type 14 (SCA14), as a novel amyloidogenic protein with no previously characterized amyloid-prone domains. We found that overexpression of PKCγ in cultured cells, as well as in vitro incubation of PKCγ without heat or chemical denaturants, causes amyloid-like fibril formation of this protein. We also observed that SCA14-associated mutations in PKCγ accelerate the amyloid-like fibril formation both in cultured cells and in vitro. We show that the C1A and kinase domains of PKCγ are involved in its soluble dimer and aggregate formation and that SCA14-associated mutations in the C1 domain cause its misfolding and aggregation. Furthermore, long-term time-lapse imaging indicates that aggregates of mutant PKCγ are highly toxic to neuronal cells. Based on these findings, we propose that PKCγ could form amyloid-like fibrils in physiological and/or pathophysiological conditions such as SCA14. More generally, our results provide novel insights into the mechanism of amyloid-like fibril formation by multi-domain proteins.


Assuntos
Amiloide/metabolismo , Proteína Quinase C/metabolismo , Degenerações Espinocerebelares/enzimologia , Amiloide/química , Amiloide/genética , Humanos , Mutação , Ligação Proteica , Proteína Quinase C/química , Proteína Quinase C/genética , Estrutura Terciária de Proteína , Ataxias Espinocerebelares , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/metabolismo
19.
Cerebellum ; 13(3): 323-30, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24242763

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disorder caused by the expansion of a polyglutamine tract in the ataxin-1 protein. To date, no fundamental treatments for SCA1 have been elucidated. However, some studies have shown that mesenchymal stem cells (MSCs) are partially effective in other genetic mouse models of cerebellar ataxia. In this study, we tested the efficacy of the intrathecal injection of MSCs in the treatment of SCA1 in transgenic (SCA1-Tg) mice. We found that intrathecal injection of only 3 × 10(3) MSCs greatly mitigated the cerebellar neuronal disorganization observed in SCA1 transgenic mice (SCA1-Tg mice). Although the Purkinje cells (PCs) of 24-week-old nontreated SCA1-Tg mice displayed a multilayer arrangement, SCA1-Tg mice at a similar age injected with MSCs displayed monolayer PCs. Furthermore, intrathecal injection of MSCs suppressed the atrophy of PC dendrites in SCA1-Tg mice. Finally, behavioral tests demonstrated that MSCs normalized deficits in motor coordination in SCA1-Tg mice. Future studies should be performed to develop optimal protocols for intrathecal transplantation of MSCs in SCA1 model primates with the aim of developing applications for SCA1 patients.


Assuntos
Cerebelo/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Ataxias Espinocerebelares/terapia , Animais , Dendritos/metabolismo , Dendritos/patologia , Modelos Animais de Doenças , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos , Camundongos Transgênicos , Doenças Neurodegenerativas/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células de Purkinje/citologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia
20.
Cerebellum ; 13(1): 29-41, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23955261

RESUMO

Spinocerebellar ataxia type 3 (SCA3) is caused by the abnormal expansion of CAG repeats within the ataxin-3 gene. Previously, we generated transgenic mice (SCA3 mice) that express a truncated form of ataxin-3 containing abnormally expanded CAG repeats specifically in cerebellar Purkinje cells (PCs). Here, we further characterize these SCA3 mice. Whole-cell patch-clamp analysis of PCs from advanced-stage SCA3 mice revealed a significant decrease in membrane capacitance due to poor dendritic arborization and the complete absence of metabotropic glutamate receptor subtype1 (mGluR1)-mediated retrograde suppression of synaptic transmission at parallel fiber terminals, with an overall preservation of AMPA receptor-mediated fast synaptic transmission. Because these cerebellar phenotypes are reminiscent of retinoic acid receptor-related orphan receptor α (RORα)-defective staggerer mice, we examined the levels of RORα in the SCA3 mouse cerebellum by immunohistochemistry and found a marked reduction of RORα in the nuclei of SCA3 mouse PCs. To confirm that the defects in SCA3 mice were caused by postnatal deposition of mutant ataxin-3 in PCs, not by genome disruption via transgene insertion, we tried to reduce the accumulation of mutant ataxin-3 in developing PCs by viral vector-mediated expression of CRAG, a molecule that facilitates the degradation of stress proteins. Concomitant with the removal of mutant ataxin-3, CRAG-expressing PCs had greater numbers of differentiated dendrites compared to non-transduced PCs and exhibited retrograde suppression of synaptic transmission following mGluR1 activation. These results suggest that postnatal nuclear accumulation of mutant ataxin-3 disrupts dendritic differentiation and mGluR-signaling in SCA3 mouse PCs, and this disruption may be caused by a defect in a RORα-driven transcription pathway.


Assuntos
Cerebelo/fisiologia , Dendritos/fisiologia , Proteínas Nucleares/metabolismo , Células de Purkinje/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Fatores de Transcrição/metabolismo , Potenciais de Ação , Animais , Ataxina-3 , Núcleo Celular/fisiologia , Cerebelo/crescimento & desenvolvimento , Dendritos/patologia , Capacitância Elétrica , Técnicas In Vitro , Doença de Machado-Joseph/genética , Doença de Machado-Joseph/patologia , Doença de Machado-Joseph/fisiopatologia , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Peptídeos , Células de Purkinje/patologia , Receptores de AMPA/metabolismo , Transmissão Sináptica , Fatores de Transcrição/química , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA