Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
EMBO J ; 42(11): e110384, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37083045

ABSTRACT

Most adult hippocampal neural stem cells (NSCs) remain quiescent, with only a minor portion undergoing active proliferation and neurogenesis. The molecular mechanisms that trigger the transition from quiescence to activation are still poorly understood. Here, we found the activity of the transcriptional co-activator Yap1 to be enriched in active NSCs. Genetic deletion of Yap1 led to a significant reduction in the relative proportion of active NSCs, supporting a physiological role of Yap1 in regulating the transition from quiescence to activation. Overexpression of wild-type Yap1 in adult NSCs did not induce NSC activation, suggesting tight upstream control mechanisms, but overexpression of a gain-of-function mutant (Yap1-5SA) elicited cell cycle entry in NSCs and hilar astrocytes. Consistent with a role of Yap1 in NSC activation, single cell RNA sequencing revealed a partial induction of an activated NSC gene expression program. Furthermore, Yap1-5SA expression also induced expression of Taz and other key components of the Yap/Taz regulon that were previously identified in glioblastoma stem cell-like cells. Consequently, dysregulated Yap1 activity led to repression of hippocampal neurogenesis, aberrant cell differentiation, and partial acquisition of a glioblastoma stem cell-like signature.


Subject(s)
Glioblastoma , Neural Stem Cells , Adult , Humans , Glioblastoma/metabolism , Cell Differentiation/physiology , Hippocampus/metabolism , Neurogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Neural Stem Cells/metabolism
2.
Nat Commun ; 12(1): 4335, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267208

ABSTRACT

Astrocytes have essential functions in brain homeostasis that are established late in differentiation, but the mechanisms underlying the functional maturation of astrocytes are not well understood. Here we identify extensive transcriptional changes that occur during murine astrocyte maturation in vivo that are accompanied by chromatin remodelling at enhancer elements. Investigating astrocyte maturation in a cell culture model revealed that in vitro-differentiated astrocytes lack expression of many mature astrocyte-specific genes, including genes for the transcription factors Rorb, Dbx2, Lhx2 and Fezf2. Forced expression of these factors in vitro induces distinct sets of mature astrocyte-specific transcripts. Culturing astrocytes in a three-dimensional matrix containing FGF2 induces expression of Rorb, Dbx2 and Lhx2 and improves astrocyte maturity based on transcriptional and chromatin profiles. Therefore, extrinsic signals orchestrate the expression of multiple intrinsic regulators, which in turn induce in a modular manner the transcriptional and chromatin changes underlying astrocyte maturation.


Subject(s)
Astrocytes/cytology , Astrocytes/physiology , Chromatin/genetics , Transcription Factors/genetics , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cerebral Cortex/cytology , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing , Epigenesis, Genetic , Gene Expression , Male , Mice, Inbred C57BL , Single-Cell Analysis , Transcription Factors/metabolism
3.
Sci Transl Med ; 11(506)2019 08 21.
Article in English | MEDLINE | ID: mdl-31434754

ABSTRACT

Niemann-Pick disease type A (NPD-A) is a lysosomal storage disorder characterized by neurodegeneration and early death. It is caused by loss-of-function mutations in the gene encoding for acid sphingomyelinase (ASM), which hydrolyzes sphingomyelin into ceramide. Here, we evaluated the safety of cerebellomedullary (CM) cistern injection of adeno-associated viral vector serotype 9 encoding human ASM (AAV9-hASM) in nonhuman primates (NHP). We also evaluated its therapeutic benefit in a mouse model of the disease (ASM-KO mice). We found that CM injection in NHP resulted in widespread transgene expression within brain and spinal cord cells without signs of toxicity. CM injection in the ASM-KO mouse model resulted in hASM expression in cerebrospinal fluid and in different brain areas without triggering an inflammatory response. In contrast, direct cerebellar injection of AAV9-hASM triggered immune response. We also identified a minimally effective therapeutic dose for CM injection of AAV9-hASM in mice. Two months after administration, the treatment prevented motor and memory impairment, sphingomyelin (SM) accumulation, lysosomal enlargement, and neuronal death in ASM-KO mice. ASM activity was also detected in plasma from AAV9-hASM CM-injected ASM-KO mice, along with reduced SM amount and decreased inflammation in the liver. Our results support CM injection for future AAV9-based clinical trials in NPD-A as well as other lysosomal storage brain disorders.


Subject(s)
Dependovirus/metabolism , Genetic Therapy , Niemann-Pick Disease, Type A/genetics , Niemann-Pick Disease, Type A/therapy , Serogroup , Animals , Brain/metabolism , Brain/pathology , Humans , Inflammation/pathology , Injections , Liver/pathology , Mice, Knockout , Motor Activity , Primates , Sphingomyelin Phosphodiesterase/administration & dosage , Sphingomyelin Phosphodiesterase/blood , Sphingomyelin Phosphodiesterase/genetics , Transgenes
4.
Front Cell Dev Biol ; 7: 62, 2019.
Article in English | MEDLINE | ID: mdl-31080799

ABSTRACT

In rodents, the hippocampal dentate gyrus gives rise to newly generated dentate granule cells (DGCs) throughout life. This process, named adult hippocampal neurogenesis (AHN), converges in the functional integration of mature DGCs into the trisynaptic hippocampal circuit. Environmental enrichment (EE) is one of the most potent positive regulators of AHN. This paradigm includes the combination of three major stimulatory components, namely increased physical activity, constant cognitive stimulation, and higher social interaction. In this regard, the pro-neurogenic effects of physical activity and cognitive stimulation have been widely addressed in adult rodents. However, the pro-neurogenic potential of the social aspect of EE has been less explored to date. Here we tackled this question by specifically focusing on the effects of a prolonged period of social enrichment (SE) in adult female C57BL6 mice. To this end, 7-week-old mice were housed in groups of 12 per cage for 8 weeks. These mice were compared with others housed under control housing (2-3 mice per cage) or EE (12 mice per cage plus running wheels and toys) conditions during the same period. We analyzed the number and morphology of Doublecortin-expressing (DCX+) cells. Moreover, using RGB retroviruses that allowed the labeling of three populations of newborn DGCs of different ages in the same mouse, we performed morphometric, immunohistochemical, and behavioral determinations. Both SE and EE increased the number and maturation of DCX+ cells, and caused an increase in dendritic maturation in certain populations of newborn DGCs. Moreover, both manipulations increased exploratory behavior in the Social Interaction test. Unexpectedly, our data revealed the potent neurogenesis-stimulating potential of SE in the absence of any further cognitive stimulation or increase in physical activity. Given that an increase in physical activity is strongly discouraged under certain circumstances, our findings may be relevant in the context of enhancing AHN via physical activity-independent mechanisms.

5.
Brain Res ; 1712: 47-54, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30711399

ABSTRACT

We have studied the consequences of in vivo GSK3ß overexpression in the cerebellum using transgenic mice with conditional expression where the transactivator tTA protein expression is driven by GFAP promoter. We demonstrate an increase in GSK3ß in Bergmann cells. To study cerebellar dysfunctions and evaluate motor coordination we analysed the latency to fall in the accelerating rotarod test. GSK3ß transgenic mice performed significantly better than wild-type mice and transgene shutdown with doxycycline normalizes the values in latency to fall in rotarod test. We had previously demonstrated using the same transgenic model, that overexpression of GSK3ß in the hippocampus results in an increase in neural precursor cells. However, we did not observe that increase in the number of Sox2+ cells in the cerebellum. All the same, we observed an increase in cerebellar glutamate transporters GLT1 and GLAST. These data show that GSK3ß can be a crucial kinase in cerebellum and especially in Bergmann glial cells.


Subject(s)
Glial Fibrillary Acidic Protein/genetics , Glycogen Synthase Kinase 3 beta/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Cerebellum/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glycogen Synthase Kinase 3 beta/biosynthesis , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Activity/physiology , Neural Stem Cells/metabolism , Neuroglia/metabolism , Promoter Regions, Genetic , Rotarod Performance Test/methods , Trans-Activators/metabolism
6.
Nat Commun ; 9(1): 473, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382840

ABSTRACT

The original version of this Article contained an error in the spelling of the author Álvaro Sebastián-Serrano, which was incorrectly given as Álvaro Sebastián Serrano. This has now been corrected in both the PDF and HTML versions of the Article.

7.
Front Mol Neurosci ; 11: 442, 2018.
Article in English | MEDLINE | ID: mdl-30618601

ABSTRACT

Prolonged seizures (status epilepticus, SE) may drive hippocampal dysfunction and epileptogenesis, at least partly, through an elevation in neurogenesis, dysregulation of migration and aberrant dendritic arborization of newly-formed neurons. MicroRNA-22 was recently found to protect against the development of epileptic foci, but the mechanisms remain incompletely understood. Here, we investigated the contribution of microRNA-22 to SE-induced aberrant adult neurogenesis. SE was induced by intraamygdala microinjection of kainic acid (KA) to model unilateral hippocampal neuropathology in mice. MicroRNA-22 expression was suppressed using specific oligonucleotide inhibitors (antagomir-22) and newly-formed neurons were visualized using the thymidine analog iodo-deoxyuridine (IdU) and a green fluorescent protein (GFP)-expressing retrovirus to visualize the dendritic tree and synaptic spines. Using this approach, we quantified differences in the rate of neurogenesis and migration, the structure of the apical dendritic tree and density and morphology of dendritic spines in newly-formed neurons.SE resulted in an increased rate of hippocampal neurogenesis, including within the undamaged contralateral dentate gyrus (DG). Newly-formed neurons underwent aberrant migration, both within the granule cell layer and into ectopic sites. Inhibition of microRNA-22 exacerbated these changes. The dendritic diameter and the density and average volume of dendritic spines were unaffected by SE, but these parameters were all elevated in mice in which microRNA-22 was suppressed. MicroRNA-22 inhibition also reduced the length and complexity of the dendritic tree, independently of SE. These data indicate that microRNA-22 is an important regulator of morphogenesis of newly-formed neurons in adults and plays a role in supressing aberrant neurogenesis associated with SE.

8.
Nat Commun ; 8(1): 2275, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273751

ABSTRACT

Excitotoxicity, a critical process in neurodegeneration, induces oxidative stress and neuronal death through mechanisms largely unknown. Since oxidative stress activates protein kinase D1 (PKD1) in tumor cells, we investigated the effect of excitotoxicity on neuronal PKD1 activity. Unexpectedly, we find that excitotoxicity provokes an early inactivation of PKD1 through a dephosphorylation-dependent mechanism mediated by protein phosphatase-1 (PP1) and dual specificity phosphatase-1 (DUSP1). This step turns off the IKK/NF-κB/SOD2 antioxidant pathway. Neuronal PKD1 inactivation by pharmacological inhibition or lentiviral silencing in vitro, or by genetic inactivation in neurons in vivo, strongly enhances excitotoxic neuronal death. In contrast, expression of an active dephosphorylation-resistant PKD1 mutant potentiates the IKK/NF-κB/SOD2 oxidative stress detoxification pathway and confers neuroprotection from in vitro and in vivo excitotoxicity. Our results indicate that PKD1 inactivation underlies excitotoxicity-induced neuronal death and suggest that PKD1 inactivation may be critical for the accumulation of oxidation-induced neuronal damage during aging and in neurodegenerative disorders.


Subject(s)
Cell Death , Neurons/metabolism , Neuroprotection , Oxidative Stress , Protein Kinase C/metabolism , Animals , Dual Specificity Phosphatase 1/metabolism , I-kappa B Kinase/metabolism , In Vitro Techniques , Mice , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism , Signal Transduction , Superoxide Dismutase/metabolism
9.
Mol Neurodegener ; 12(1): 59, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28810892

ABSTRACT

BACKGROUND: Extracellular Tau is toxic for neighboring cells, and it contributes to the progression of AD. The CX3CL1/CX3CR1 axis is an important neuron/microglia communication mechanism. METHODS: We studied Tau clearance by microglia both in vitro (microglia primary cultures treated with Cy5-Tau, affinity chromatography to study the binding of Tau to CX3CR1, and Tau-CX3CL1 competition assays) and in vivo (stereotaxic injection of Cy5-Tau into WT and CX3CR1-/- mice). The expression of CX3CR1, CX3CL1 and the microglial phagocytic phenotype were studied in brain tissue samples from AD patients. RESULTS: Tau binding to CX3CR1 triggers the internalization of the former by microglia, whereas S396 Tau phosphorylation decreases the binding affinity of this protein to CX3CR1. Of note, the progressive increase in the levels of phosho-Tau occurred in parallel with an increase in CX3CR1. In addition, our studies suggest that the phagocytic capacity of microglia in brain tissue samples from AD patients is decreased. Furthermore, the CX3CR1/CX3CL1 axis may be impaired in late stages of the disease. CONCLUSIONS: Our data suggest that the CX3CR1/CX3CL1 axis plays a key role in the phagocytosis of Tau by microglia in vitro and in vivo and that it is affected as AD progresses. Taken together, our results reveal CX3CR1 as a novel target for the clearance of extracellular Tau.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , CX3C Chemokine Receptor 1/metabolism , Microglia/metabolism , tau Proteins/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , Mice, Knockout , Neurons/metabolism , Phosphorylation
10.
Mol Cell ; 66(3): 358-372.e7, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28475871

ABSTRACT

A decline in proteasome function is causally connected to neuronal aging and aging-associated neuropathologies. By using hippocampal neurons in culture and in vivo, we show that aging triggers a reduction and a cytoplasm-to-nucleus redistribution of the E3 ubiquitin ligase mahogunin (MGRN1). Proteasome impairment induces MGRN1 monoubiquitination, the key post-translational modification for its nuclear entry. One potential mechanism for MGRN1 monoubiquitination is via progressive deubiquitination at the proteasome of polyubiquitinated MGRN1. Once in the nucleus, MGRN1 potentiates the transcriptional cellular response to proteotoxic stress. Inhibition of MGRN1 impairs ATF3-mediated neuronal responsiveness to proteosomal stress and increases neuronal stress, while increasing MGRN1 ameliorates signs of neuronal aging, including cognitive performance in old animals. Our results imply that, among others, the strength of neuronal survival in a proteasomal deterioration background, like during aging, depends on the fine-tuning of ubiquitination-deubiquitination.


Subject(s)
Aging/metabolism , Cell Nucleus/enzymology , Cytoplasm/enzymology , Hippocampus/enzymology , Neurons/enzymology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitination , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Active Transport, Cell Nucleus , Aging/genetics , Aging/pathology , Animals , Behavior, Animal , Cell Nucleus/ultrastructure , Cell Survival , Chromatin/enzymology , Cognition , HEK293 Cells , Hippocampus/ultrastructure , Humans , Maze Learning , Mice, Inbred C57BL , Neurons/ultrastructure , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Rats, Wistar , Signal Transduction , Stress, Physiological , Transcription, Genetic , Transfection , Ubiquitin-Protein Ligases/genetics
11.
Cell Mol Life Sci ; 74(6): 1153-1163, 2017 03.
Article in English | MEDLINE | ID: mdl-27832289

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß) is a serine-threonine kinase implicated in multiple processes and signaling pathways. Its dysregulation is associated with different pathological conditions including Alzheimer's disease (AD). Here we demonstrate how changes in GSK-3ß activity and/or levels regulate the production and subsequent secretion of fractalkine, a chemokine involved in the immune response that has been linked to AD and to other different neurological disorders. Treatment of primary cultured neurons with GSK-3ß inhibitors such as lithium and AR-A014418 decreased full-length fractalkine in total cell extracts. Opposite effects were observed after neuron transduction with a lentiviral vector overexpressing the kinase. Biotinylation assays showed that those changes mainly affect the plasma membrane-associated form of the protein, an observation that positively correlates with changes in the levels of its soluble form. These effects were confirmed in lithium-treated wild type (wt) mice and in GSK-3ß transgenic animals, as well as in brain samples from AD patients, evident as age-dependent (animals) or Braak stage dependent changes (humans) in both the membrane-bound and the soluble forms of the protein. Further immunohistochemical analyses demonstrated how GSK-3ß exerts these effects by affecting the trafficking of the chemokine from the Golgi to the plasma membrane, in different and opposite ways when the levels/activity of the kinase are increased or decreased. This work provides for the first time a mechanism linking GSK-3ß and fractalkine both in vitro and in vivo, with important implications for neurological disorders and especially for AD, in which levels of this chemokine might be useful as a diagnostic tool.


Subject(s)
Alzheimer Disease/metabolism , Cell Membrane/metabolism , Chemokine CX3CL1/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Golgi Apparatus/metabolism , Animals , Humans , Mice, Inbred C57BL , Protein Binding , Protein Transport , Solubility , Transport Vesicles/metabolism , rab GTP-Binding Proteins/metabolism
12.
EMBO J ; 35(13): 1417-36, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27198172

ABSTRACT

Tau is a microtubule-associated neuronal protein found mainly in axons. However, its presence in dendrites and dendritic spines is particularly relevant due to its involvement in synaptic plasticity and neurodegeneration. Here, we show that Tau plays a novel in vivo role in the morphological and synaptic maturation of newborn hippocampal granule neurons under basal conditions. Furthermore, we reveal that Tau is involved in the selective cell death of immature granule neurons caused by acute stress. Also, Tau deficiency protects newborn neurons from the stress-induced dendritic atrophy and loss of postsynaptic densities (PSDs). Strikingly, we also demonstrate that Tau regulates the increase in newborn neuron survival triggered by environmental enrichment (EE). Moreover, newborn granule neurons from Tau(-/-) mice did not show any stimulatory effect of EE on dendritic development or on PSD generation. Thus, our data demonstrate that Tau(-/-) mice show impairments in the maturation of newborn granule neurons under basal conditions and that they are insensitive to the modulation of adult hippocampal neurogenesis exerted by both stimulatory and detrimental stimuli.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Neurogenesis , tau Proteins/metabolism , Animals , Mice , Mice, Knockout
13.
Cell Mol Life Sci ; 73(18): 3569-82, 2016 09.
Article in English | MEDLINE | ID: mdl-27010990

ABSTRACT

Adult hippocampal neurogenesis (AHN) is a key process for certain types of hippocampal-dependent learning. Alzheimer's disease (AD) is accompanied by memory deficits related to alterations in AHN. Given that the increased activity of GSK-3ß has been related to alterations in the population of hippocampal granule neurons in AD patients, we designed a novel methodology by which to induce selective GSK-3ß overexpression exclusively in newborn granule neurons. To this end, we injected an rtTA-IRES-EGFP-expressing retrovirus into the hippocampus of tTO-GSK-3ß mice. Using this novel retroviral strategy, we found that GSK-3ß caused a cell-autonomous impairment of the morphological and synaptic maturation of newborn neurons. In addition, we examined whether GSK-3ß overexpression in newborn neurons limits the effects of physical activity. While physical exercise increased the number of dendritic spines, the percentage of mushroom spines, and the head diameter of the same in tet-OFF cells, these effects were not triggered in tet-ON cells. This observation suggests that GSK-3ß blocks the stimulatory actions of exercise. Given that the activity of GSK-3ß is increased in the brains of individuals with AD, these data may be relevant for non-pharmacological therapies for AD.


Subject(s)
Genetic Vectors/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Neurons/metabolism , Physical Conditioning, Animal , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Female , Genetic Vectors/genetics , Glycogen Synthase Kinase 3 beta/genetics , Immunohistochemistry , Mice , Microscopy, Fluorescence , Neurogenesis , Phosphorylation , Retroviridae/genetics , Spine/physiology , tau Proteins/metabolism
14.
J Biol Chem ; 291(15): 8199-213, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26887949

ABSTRACT

In restricted areas of the adult brain, like the subgranular zone of the dentate gyrus (DG), there is continuous production of new neurons. This process, named adult neurogenesis, is involved in important cognitive functions such as memory and learning. It requires the presence of newborn neurons that arise from neuronal stem cells, which divide and differentiate through successive stages in adulthood. In this work, we demonstrate that overexpression of glycogen synthase kinase (GSK) 3ß in neural precursor cells (NPCs) using the glial fibrillary acidic protein promoter during DG development produces an increase in the neurogenic process, increasing NPCs numbers. Moreover, the transgenic mice show higher DG volume and increased number of mature granule neurons. In an attempt to compensate for these alterations, glial fibrillary acidic protein/GSK3ß-overexpressing mice show increased levels of Dkk1 and sFRP3, two inhibitors of the Wnt-frizzled complex. We have also found behavioral differences between wild type and transgenic mice, indicating a higher rating in memory tasks for GSK3ß-overexpressing mice compared with wild type mice. These data indicate that GSK3ß is a crucial kinase in NPC physiology and suggest that this molecule plays a key role in the correct development of DG and adult neurogenesis in this region.


Subject(s)
Dentate Gyrus/growth & development , Glycogen Synthase Kinase 3/genetics , Memory , Neural Stem Cells/metabolism , Neurogenesis , Up-Regulation , Animals , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Glial Fibrillary Acidic Protein/genetics , Glycogen Synthase Kinase 3 beta , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Promoter Regions, Genetic
15.
Brain Behav Immun ; 53: 242-254, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26724574

ABSTRACT

Recent experimental data suggest that mood disorders are related to inflammatory phenomena and have led to the "inflammatory hypothesis of depression". Given that the hippocampus is one of the most affected areas in these disorders, we used a model of acute stress (the Porsolt test) to evaluate the consequences of forced swimming on two crucial events related to the pathophysiology of major depression: the functional maturation of newborn granule neurons; and the hippocampal inflammatory milieu. Using PSD95:GFP-expressing retroviruses, we found that forced swimming selectively alters the dendritic morphology of newborn neurons and impairs their connectivity by reducing the number and volume of their postsynaptic densities. In addition, acute stress triggered a series of morphological changes in microglial cells, together with an increase in microglial CD68 expression, thus suggesting the functional and morphological activation of this cell population. Furthermore, we observed an intriguing change in the hippocampal inflammatory milieu in response to forced swimming. Importantly, the levels of several molecules affected by acute stress (such as Interleukin-6 and eotaxin) have been described to also be altered in patients with depression and other mood disorders.


Subject(s)
Neurogenesis/physiology , Neurons/physiology , Stress, Physiological/physiology , Animals , Dendrites/metabolism , Dendrites/physiology , Depression/metabolism , Depression/pathology , Depressive Disorder, Major/metabolism , Female , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Inbred BALB C , Microglia/metabolism , Microglia/pathology , Models, Animal , Neurons/metabolism , Swimming
16.
J Alzheimers Dis ; 50(1): 77-87, 2016.
Article in English | MEDLINE | ID: mdl-26638867

ABSTRACT

The microtubule-associated protein (MAP) tau plays a critical role in the pathogenesis of tauopathies. Excess tau can be released into the extracellular medium in a physiological or pathological manner to be internalized by surrounding neurons-a process that contributes to the spread of this protein throughout the brain. Such spreading may correlate with the progression of the abovementioned diseases. In addition to neurons, tau can be internalized into other cells. Here we demonstrate that microglia take up tau in vitro and in vivo. In this regard, microglia from primary cultures internalized soluble (human recombinant tau42) and insoluble (homogenates derived from human AD brain) tau in vitro. Furthermore, using stereotaxic injection of tau in mice in vivo, we show that murine microglia internalize human tau. In addition, we demonstrate, for the first time, that microglia colocalize with various forms of tau in postmortem brain tissue of patients with Alzheimer's disease and non-demented control subjects. Our data reveal a potential role of microglia in the internalization of tau that might be relevant for the design of strategies to enhance the clearance of extracellular tau in neurodegenerative diseases characterized by the accumulation of this protein.


Subject(s)
Alzheimer Disease/pathology , Microglia/metabolism , tau Proteins/metabolism , Analysis of Variance , Animals , Animals, Newborn , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phosphorylation , Protein Transport/physiology , Rats , Time Factors
17.
Brain Res ; 1611: 84-92, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-25779040

ABSTRACT

In neuronal cultures, glycogen synthase kinase 3(GSK3) is truncated at the N-terminal end by calpain downstream of activated glutamate receptors. However, the in vivo biological significance of that truncation has not been explored. In an attempt to elucidate if GSK3 truncation has a pathophysiological relevance, we have used intraperitoneal injections of kainic acid (KA) in rats and intra-amygdala KA microinjections in mice as in vivo models of excitotoxicity. Spectrin cleavage analyzed by immunohistochemistry was observed in the CA1 hippocampal field in KA-intraperitoneal treated rats while the CA3 region was the hippocampal area affected after intra-amygdala KA microinjections. GSK3ß immunofluorescence did not colocalize with truncated spectrin in both treatments using an antibody that recognize the N-terminal end of GSK3ß. Thus, those neurons which are spectrin-positive do not show GSK3ß immunolabelling. To study GSK3ß truncation in vitro, we exposed organotypic hippocampal slices and cultured cortical neurons to KA leading to the truncation of GSK3 and we found that truncation was blocked by the calpain inhibitor calpeptin. These data suggest a relationship between N-terminal GSK3ß truncation and excitotoxicity. Overall, our data reinforces the important relationship between glutamate receptors and GSK3 and their role in neurodegenerative processes in which excitotoxicity is involved.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Glycogen Synthase Kinase 3/metabolism , Hippocampus/enzymology , Kainic Acid/toxicity , Neurons/enzymology , Amygdala/drug effects , Animals , Antibodies , Cells, Cultured , Disease Models, Animal , Glycogen Synthase Kinase 3/immunology , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Rats , Rats, Wistar , Spectrin/metabolism
18.
Exp Neurol ; 263: 285-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446721

ABSTRACT

Newborn neurons are continuously added to the hippocampal dentate gyrus (DG) throughout life. Mature and immature granule neurons are believed to send their axonal projections exclusively to the hippocampal CA3 field. However, recent data point to an alternative trisynaptic circuit, involving a direct axonal projection from mature granule neurons to the CA2 field. Whether this circuit takes place only in mature granule neurons or, on the contrary, whether immature granule neurons also contribute to this novel connection is unknown. We used various retroviral vectors to show that immature granule neurons send axonal processes to and establish synaptic contacts with CA2 pyramidal neurons and that axonal growth follows a similar time course to that described for CA3 innervation. In addition, we provide experimental evidence demonstrating that the pathway connecting newborn granule neurons and the CA2 field can be modulated by physiological and deleterious stimuli.


Subject(s)
CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/growth & development , Neural Pathways/growth & development , Neurogenesis/physiology , Neurons/cytology , Animals , Animals, Newborn , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neural Pathways/cytology
19.
J Alzheimers Dis ; 40(1): 143-51, 2014.
Article in English | MEDLINE | ID: mdl-24366920

ABSTRACT

We modified tau protein with boronic acid to facilitate its delivery into non neural or neural cultured cells lacking tau protein. Our results indicate that the incorporated tau promotes the formation of cytoplasmic extensions in non-neuronal cells, as well as the appearance of neurites in cultured tau knockout hippocampal neurons. In addition, boronated tau is incorporated into hippocampal neurons of tau knockout mice after intracranial injection in vivo. These findings describe a novel method to deliver exogenous tau protein into cells.


Subject(s)
Neurons/metabolism , tau Proteins/metabolism , Animals , Boron Compounds/metabolism , Boron Compounds/pharmacology , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian , Hippocampus/cytology , Hippocampus/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/ultrastructure , Tetrahydronaphthalenes/metabolism , Tetrahydronaphthalenes/pharmacology , Tubulin/metabolism , tau Proteins/genetics
20.
Hum Mol Genet ; 22(3): 466-82, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23118350

ABSTRACT

Failures in neurotrophic support and signalling play key roles in Alzheimer's disease (AD) pathogenesis. We previously demonstrated that downregulation of the neurotrophin effector Kinase D interacting substrate (Kidins220) by excitotoxicity and cerebral ischaemia contributed to neuronal death. This downregulation, triggered through overactivation of N-methyl-D-aspartate receptors (NMDARs), involved proteolysis of Kidins220 by calpain and transcriptional inhibition. As excitotoxicity is at the basis of AD aetiology, we hypothesized that Kidins220 might also be downregulated in this disease. Unexpectedly, Kidins220 is augmented in necropsies from AD patients where it accumulates with hyperphosphorylated tau. This increase correlates with enhanced Kidins220 resistance to calpain processing but no higher gene transcription. Using AD brain necropsies, glycogen synthase kinase 3-ß (GSK3ß)-transgenic mice and cell models of AD-related neurodegeneration, we show that GSK3ß phosphorylation decreases Kidins220 susceptibility to calpain proteolysis, while protein phosphatase 1 (PP1) action has the opposite effect. As altered activities of GSK3ß and phosphatases are involved in tau aggregation and constitute hallmarks in AD, a GSK3ß/PP1 imbalance may also contribute to Kidins220 decreased clearance, accumulation and hampered neurotrophin signalling from early stages of the disease pathogenesis. These results encourage searches for mutations in Kidins220 gene and their possible associations to dementias. Finally, our data support a model where the effects of excitotoxicity drastically differ when occurring in cerebral ischaemia versus progressively sustained toxicity along AD progression. The striking differences in Kidins220 stability resulting from chronic versus acute brain damage may also have important implications for the therapeutic intervention of neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Calpain/metabolism , Glycogen Synthase Kinase 3/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Phosphatase 1/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Calpain/genetics , Cell Death , Cells, Cultured , Disease Models, Animal , Down-Regulation , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/pathology , Okadaic Acid/adverse effects , Phosphorylation , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/genetics , Proteolysis , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...