Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Neurobiol Dis ; 181: 106106, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37001613

ABSTRACT

Epilepsy is a comorbidity associated with Alzheimer's disease (AD), often starting many years earlier than memory decline. Investigating this association in the early pre-symptomatic stages of AD can unveil new mechanisms of the pathology as well as guide the use of antiepileptic drugs to prevent or delay hyperexcitability-related pathological effects of AD. We investigated the impact of repeated seizures on hippocampal memory and amyloid-ß (Aß) load in pre-symptomatic Tg2576 mice, a transgenic model of AD. Seizure induction caused memory deficits and an increase in oligomeric Aß42 and fibrillary species selectively in pre-symptomatic transgenic mice, and not in their wildtype littermates. Electrophysiological patch-clamp recordings in ex vivo CA1 pyramidal neurons and immunoblots were carried out to investigate the neuronal alterations associated with the behavioral outcomes of Tg2576 mice. CA1 pyramidal neurons exhibited increased intrinsic excitability and lower hyperpolarization-activated Ih current. CA1 also displayed lower expression of the hyperpolarization-activated cyclic nucleotide-gated HCN1 subunit, a protein already identified as downregulated in the AD human proteome. The antiepileptic drug lamotrigine restored electrophysiological alterations and prevented both memory deficits and the increase in extracellular Aß induced by seizures. Thus our study provides evidence of pre-symptomatic hippocampal neuronal alterations leading to hyperexcitability and associated with both higher susceptibility to seizures and to AD-specific seizure-induced memory impairment. Our findings also provide a basis for the use of the antiepileptic drug lamotrigine as a way to counteract acceleration of AD induced by seizures in the early phases of the pathology.


Subject(s)
Alzheimer Disease , Mice , Humans , Animals , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Anticonvulsants/pharmacology , Lamotrigine/adverse effects , Hippocampus/metabolism , Amyloid beta-Peptides/metabolism , Seizures/pathology , Mice, Transgenic , Disease Models, Animal , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/prevention & control
2.
Neural Plast ; 2022: 9959044, 2022.
Article in English | MEDLINE | ID: mdl-35075360

ABSTRACT

The mammalian hippocampal dentate gyrus is a niche for adult neurogenesis from neural stem cells. Newborn neurons integrate into existing neuronal networks, where they play a key role in hippocampal functions, including learning and memory. In the ageing brain, neurogenic capability progressively declines while in parallel increases the risk for developing Alzheimer's disease (AD), the main neurodegenerative disorder associated with memory loss. Numerous studies have investigated whether impaired adult neurogenesis contributes to memory decline in AD. Here, we review the literature on adult hippocampal neurogenesis (AHN) and AD by focusing on both human and mouse model studies. First, we describe key steps of AHN, report recent evidence of this phenomenon in humans, and describe the specific contribution of newborn neurons to memory, as evinced by animal studies. Next, we review articles investigating AHN in AD patients and critically examine the discrepancies among different studies over the last two decades. Also, we summarize researches investigating AHN in AD mouse models, and from these studies, we extrapolate the contribution of molecular factors linking AD-related changes to impaired neurogenesis. Lastly, we examine animal studies that link impaired neurogenesis to specific memory dysfunctions in AD and review treatments that have the potential to rescue memory capacities in AD by stimulating AHN.


Subject(s)
Alzheimer Disease/physiopathology , Hippocampus/physiopathology , Memory/physiology , Neurogenesis/physiology , Animals , Humans , Neural Stem Cells/physiology , Neurons/physiology
3.
Pharmacol Res ; 172: 105795, 2021 10.
Article in English | MEDLINE | ID: mdl-34339837

ABSTRACT

Neuroinflammation can severely affect brain homeostasis and adult hippocampal neurogenesis with detrimental effects on cognitive processes. Brain and gut are intimately connected via the "gut-brain axis", a bidirectional communication system, and the administration of live bacteria (probiotics) has been shown to represent an intriguing approach for the prevention or even the cure of several diseases. In the present study we evaluated the putative neuroprotective effect of 15-days consumption of a multi-strain probiotic formulation based on food-associated strains and human gut bacteria at the dose of 109 CFU/mouse/day in a mouse model of acute inflammation, induced by an intraperitoneal single injection of LPS (0.1 mg/kg) at the end of probiotic administration. The results indicate that the prolonged administration of the multi-strain probiotic formulation not only prevents the LPS-dependent increase of pro-inflammatory cytokines in specific regions of the brain (hippocampus and cortex) and in the gastrointestinal district but also triggers a potent proneurogenic response capable of enhancing hippocampal neurogenesis. This effect is accompanied by a potentiation of intestinal barrier, as documented by the increased epithelial junction expression in the colon. Our hypothesis is that pre-treatment with the multi-strain probiotic formulation helps to create a systemic protection able to counteract or alleviate the effects of LPS-dependent acute pro-inflammatory responses.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain-Gut Axis , Neuroinflammatory Diseases/prevention & control , Neuroprotective Agents/therapeutic use , Probiotics , Animals , Anxiety , Brain/cytology , Cadherins/metabolism , Colon/metabolism , Cytokines/genetics , Disease Models, Animal , Exploratory Behavior , Illness Behavior , Lipopolysaccharides , Male , Mice, Inbred C57BL , Neurogenesis , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/microbiology , Occludin/metabolism
4.
J Neurosci ; 36(20): 5437-47, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194325

ABSTRACT

UNLABELLED: Phosphorylation of serine/threonine residues preceding a proline regulates the fate of its targets through postphosphorylation conformational changes catalyzed by the peptidyl-prolyl cis-/trans isomerase Pin1. By flipping the substrate between two different functional conformations, this enzyme exerts a fine-tuning of phosphorylation signals. Pin1 has been detected in dendritic spines and shafts where it regulates protein synthesis required to sustain the late phase of long-term potentiation (LTP). Here, we demonstrate that Pin1 residing in postsynaptic structures can interact with postsynaptic density protein-95 (PSD-95), a key scaffold protein that anchors NMDA receptors (NMDARs) in PSD via GluN2-type receptor subunits. Pin1 recruitment by PSD-95 occurs at specific serine-threonine/proline consensus motifs localized in the linker region connecting PDZ2 to PDZ3 domains. Upon binding, Pin1 triggers structural changes in PSD-95, thus negatively affecting its ability to interact with NMDARs. In electrophysiological experiments, larger NMDA-mediated synaptic currents, evoked in CA1 principal cells by Schaffer collateral stimulation, were detected in hippocampal slices obtained from Pin1(-/-) mice compared with controls. Similar results were obtained in cultured hippocampal cells expressing a PSD-95 mutant unable to undergo prolyl-isomerization, thus indicating that the action of Pin1 on PSD-95 is critical for this effect. In addition, an enhancement in spine density and size was detected in CA1 principal cells of Pin1(-/-) or in Thy-1GFP mice treated with the pharmacological inhibitor of Pin1 catalytic activity PiB.Our data indicate that Pin1 controls synaptic content of NMDARs via PSD-95 prolyl-isomerization and the expression of dendritic spines, both required for LTP maintenance. SIGNIFICANCE STATEMENT: PSD-95, a membrane-associated guanylate kinase, is the major scaffolding protein at excitatory postsynaptic densities and a potent regulator of synaptic strength and plasticity. The activity of PSD-95 is tightly controlled by several post-translational mechanisms including proline-directed phosphorylation. This signaling cascade regulates the fate of its targets through postphosphorylation conformational modifications catalyzed by the peptidyl-prolyl cis-/trans isomerase Pin1. Here, we uncover a new role of Pin1 in glutamatergic signaling. By interacting with PSD-95, Pin1 dampens PSD-95 ability to complex with NMDARs, thus negatively affecting NMDAR signaling and spine morphology. Our findings further emphasize the emerging role of Pin1 as a key modulator of synaptic transmission.


Subject(s)
Guanylate Kinases/metabolism , Membrane Proteins/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , CA1 Region, Hippocampal/cytology , Cells, Cultured , Disks Large Homolog 4 Protein , Female , HEK293 Cells , Humans , Isomerism , Long-Term Potentiation , Male , Mice , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Protein Binding , Synapses/physiology , Synaptic Transmission
5.
Neural Plast ; 2017: 5281829, 2017.
Article in English | MEDLINE | ID: mdl-29435372

ABSTRACT

Neuronal activity has a strong causal role in the production and release of the neurotoxic ß-amyloid peptide (Aß). Because of this close link, gradual accumulation of Aß into amyloid plaques has been reported in brain areas with intense neuronal activity, including cortical regions that display elevated activation at resting state. However, the link between Aß and activity is not always linear and recent studies report exceptions to the view of "more activity, more plaques." Here, we review the literature about the activity-dependent production of Aß in both human cases and AD models and focus on the evidences that brain regions with elevated convergence of synaptic connections (herein referred to as brain nodes) are particularly vulnerable to Aß accumulation. Next, we will examine data supporting the hypothesis that, since Aß is released from synaptic terminals, ß-amyloidosis can spread in AD brain by advancing through synaptically connected regions, which makes brain nodes vulnerable to Aß accumulation. Finally, we consider possible mechanisms that account for ß-amyloidosis progression through synaptically linked regions.


Subject(s)
Alzheimer Disease/metabolism , Amyloidosis/metabolism , Brain/metabolism , Neurons/metabolism , Synapses/metabolism , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Amyloidosis/complications , Animals , Humans , Plaque, Amyloid/complications , Plaque, Amyloid/metabolism , Synaptic Transmission
6.
Neural Plast ; 2015: 651469, 2015.
Article in English | MEDLINE | ID: mdl-26075101

ABSTRACT

Experience modifies synaptic connectivity through processes that involve dendritic spine rearrangements in neuronal circuits. Although cAMP response element binding protein (CREB) has a key function in spines changes, its role in activity-dependent rearrangements in brain regions of rodents interacting with the surrounding environment has received little attention so far. Here we studied the effects of vibrissae trimming, a widely used model of sensory deprivation-induced cortical plasticity, on processes associated with dendritic spine rearrangements in the barrel cortex of a transgenic mouse model of CREB downregulation (mCREB mice). We found that sensory deprivation through prolonged whisker trimming leads to an increased number of thin spines in the layer V of related barrel cortex (Contra) in wild type but not mCREB mice. In the barrel field controlling spared whiskers (Ipsi), the same trimming protocol results in a CREB-dependent enlargement of dendritic spines. Last, we demonstrated that CREB regulates structural rearrangements of synapses that associate with dynamic changes of dendritic spines. Our findings suggest that CREB plays a key role in dendritic spine dynamics and synaptic circuits rearrangements that account for new brain connectivity in response to changes in the environment.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Dendritic Spines/physiology , Neuronal Plasticity , Somatosensory Cortex/physiology , Synapses/physiology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Spines/metabolism , Mice , Mice, Transgenic , Phosphorylation , Sensory Deprivation/physiology , Somatosensory Cortex/metabolism , Synapses/metabolism
7.
Neurobiol Learn Mem ; 115: 108-15, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25132316

ABSTRACT

Memory formation requires changes in neuronal networks connectivity based on modifications in strength and number of synapses. The mechanisms driving these changes have been intensively studied, but mostly under naive conditions, i.e. in animals that have not been cognitively challenged. Better characterization of synaptic requirements supporting memory formation can emerge from studies focusing on synaptic changes in memory-encoding structures while or after the animal model is cognitively challenged. Here, with this concept in mind, we review the literature describing structural, functional and molecular alterations developing in the hippocampus when animals are asked to form memories. We also briefly discuss the interest of this approach for disclosing pathological mechanisms in memory disorders, which might otherwise not be observed in naive conditions.


Subject(s)
Learning/physiology , Neuronal Plasticity/physiology , Animals , Cognition/physiology , Hippocampus/physiology , Memory Disorders/physiopathology
8.
Hippocampus ; 23(6): 488-99, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23504989

ABSTRACT

The transcription factor cAMP response element binding protein (CREB) is a key protein implicated in memory, synaptic plasticity and structural plasticity in mammals. Whether CREB regulates the synaptic incorporation of hippocampal glutamatergic receptors under basal and learning-induced conditions remains, however, mostly unknown. Using double-transgenic mice conditionally expressing a dominant negative form of CREB (CREBS133A, mCREB), we analyzed how chronic loss of CREB function in adult hippocampal glutamatergic neurons impacts the levels of the AMPA and NMDA receptors subunits within the post-synaptic densities (PSD). In basal (naïve) conditions, we report that inhibition of CREB function was associated with a specific reduction of the AMPAR subunit GluA1 and a proportional increase in its Ser845 phosphorylated form within the PSD. These molecular alterations correlated with a reduction in AMPA receptors mEPSC frequency, with a decrease in long-term potentiation (LTP), and with an increase in long-term depression (LTD). The basal levels other major synaptic proteins (GluA2/3, GluN1, GluN2A, and PSD95) within the PSD were not affected by CREB inhibition. Blocking CREB function also impaired contextual fear conditioning (CFC) and selectively blocked the CFC-driven enhancement of GluA1 and its Ser845 phosphorylated form within the PSD, molecular changes normally observed in wild-type mice. CFC-driven enhancement of other synaptic proteins (GluA2/3, GluN1, GluN2A, and PSD95) within the PSD was not significantly perturbed by the loss of CREB function. These findings provide the first evidence that, in vivo, CREB is necessary for the specific maintenance of the GluA1 subunit within the PSD of hippocampal neurons in basal conditions and for its trafficking within the PSD during the occurrence of learning.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Learning/physiology , Protein Subunits/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, AMPA/antagonists & inhibitors
9.
Learn Mem ; 19(8): 330-6, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22815537

ABSTRACT

The modulation of synaptic strength associated with learning is post-synaptically regulated by changes in density and shape of dendritic spines. The transcription factor CREB (cAMP response element binding protein) is required for memory formation and in vitro dendritic spine rearrangements, but its role in learning-induced remodeling of neurons remains elusive. Using transgenic mice conditionally expressing a dominant-negative CREB (CREBS133A: mCREB) mutant, we found that inhibiting CREB function does not alter spine density, spine morphology, and levels of polymerized actin in naive CA1 neurons. CREB inhibition, however, impaired contextual fear conditioning and produced a learning-induced collapse of spines associated with a blockade of learning-dependent increase in actin polymerization. Blocking mCREB expression with doxycycline rescued memory and restored a normal pattern of learning-induced spines, demonstrating that CREB controls structural adaptations of neurons selectively involved in memory formation.


Subject(s)
CREB-Binding Protein/physiology , Conditioning, Psychological/physiology , Neurons/metabolism , Alanine/genetics , Analysis of Variance , Animals , Animals, Newborn , CREB-Binding Protein/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Doxycycline/pharmacology , Electroshock/adverse effects , Fear/physiology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurons/cytology , Neurons/drug effects , Neurons/ultrastructure , Phosphorylation , Serine/genetics , Silver Staining
10.
EMBO Mol Med ; 15(11): e15984, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37792911

ABSTRACT

Cell signaling is central to neuronal activity and its dysregulation may lead to neurodegeneration and cognitive decline. Here, we show that selective genetic potentiation of neuronal ERK signaling prevents cell death in vitro and in vivo in the mouse brain, while attenuation of ERK signaling does the opposite. This neuroprotective effect mediated by an enhanced nuclear ERK activity can also be induced by the novel cell penetrating peptide RB5. In vitro administration of RB5 disrupts the preferential interaction of ERK1 MAP kinase with importinα1/KPNA2 over ERK2, facilitates ERK1/2 nuclear translocation, and enhances global ERK activity. Importantly, RB5 treatment in vivo promotes neuroprotection in mouse models of Huntington's (HD), Alzheimer's (AD), and Parkinson's (PD) disease, and enhances ERK signaling in a human cellular model of HD. Additionally, RB5-mediated potentiation of ERK nuclear signaling facilitates synaptic plasticity, enhances cognition in healthy rodents, and rescues cognitive impairments in AD and HD models. The reported molecular mechanism shared across multiple neurodegenerative disorders reveals a potential new therapeutic target approach based on the modulation of KPNA2-ERK1/2 interactions.


Subject(s)
MAP Kinase Signaling System , Neuroprotection , Animals , Humans , Mice , alpha Karyopherins/pharmacology , Cognition , Phosphorylation , Signal Transduction
11.
Brain Sci ; 12(2)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35203970

ABSTRACT

Imaging in neurosciences allows for the visual representation of micro- and macro-components of the central (CNS) and peripheral (PNS) nervous systems with the intent of investigating their morphology and function, to provide diagnosis and prognosis of neurological diseases and to monitor responses to treatments [...].

12.
Biomolecules ; 12(4)2022 03 31.
Article in English | MEDLINE | ID: mdl-35454121

ABSTRACT

Epidemiological evidence indicates that stress and aversive psychological conditions can affect cancer progression, while well-being protects against it. Although a large set of studies have addressed the impact of stress on cancer, not much is known about the mechanisms that protect from cancer in healthy psychological conditions. C57BL/6J mouse pups were exposed to an environmental enrichment condition consisting of being raised until weaning by the biological lactating mother plus a non-lactating virgin female (LnL = Lactating and non-Lactating mothers). The Control group consisted of mice raised by a single lactating mother (L = Lactating). Four months after weaning, mice from LnL and L conditions were exposed to intramuscular injection of 3-methylcolantrene (3MCA), a potent tumorigenic drug, and onset and progression of 3MCA-induced fibrosarcomas were monitored over time. Pups from the LnL compared to the L group received more parental care and were more resilient to stressful events during the first week of life. In association, the onset of tumors in LnL adults was significantly delayed. At the molecular level, we observed increased levels of wild-type p53 protein in tumor samples of LnL compared to L adults and higher levels of its target p21 in healthy muscles of LnL mice compared to the L group, supporting the hypothesis of potential involvement of p53 in tumor development. Our study sustains the model that early life care protects against tumor susceptibility.


Subject(s)
Carcinogenesis , Social Environment , Tumor Suppressor Protein p53 , Animals , Female , Lactation , Mice , Mice, Inbred C57BL , Tumor Suppressor Protein p53/genetics
13.
Learn Mem ; 17(5): 236-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20404004

ABSTRACT

B6-Tg/Thy1APP23Sdz (APP23) mutant mice exhibit neurohistological hallmarks of Alzheimer's disease but show intact basal hippocampal neurotransmission and synaptic plasticity. Here, we examine whether spatial learning differently modifies the structural and electrophysiological properties of hippocampal synapses in APP23 and wild-type mice. While no genotypic difference was found in the pseudotrained mice, training elicited a stronger increase in spine density and a more rapid decay of long-term potentiation (LTP) in APP23 mutants. Thus, learning discloses mutation-related abnormalities regarding dendritic spine formation and LTP persistence, thereby suggesting that although unaltered in naïve synapses, plasticity becomes defective at the time it comes into play.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/genetics , Hippocampus/physiopathology , Learning Disabilities/etiology , Long-Term Potentiation/genetics , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Analysis of Variance , Animals , Biophysics/methods , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , Disease Models, Animal , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/genetics , Hippocampus/pathology , Humans , In Vitro Techniques , Learning Disabilities/genetics , Locomotion/genetics , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Neurons/pathology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Reaction Time/genetics , Silver Staining/methods , Time Factors
14.
Brain Sci ; 11(9)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34573265

ABSTRACT

Sarcopenia, a geriatric syndrome involving loss of muscle mass and strength, is often associated with the early phases of Alzheimer's disease (AD). Pathological hallmarks of AD including amyloid ß (Aß) aggregates which can be found in peripheral tissues such as skeletal muscle. However, not much is currently known about their possible involvement in sarcopenia. We investigated neuronal innervation in skeletal muscle of Tg2576 mice, a genetic model for Aß accumulation. We examined cholinergic innervation of skeletal muscle in adult Tg2576 and wild type mice by immunofluorescence labeling of tibialis anterior (TA) muscle sections using antibodies raised against neurofilament light chain (NFL) and acetylcholine (ACh) synthesizing enzyme choline acetyltransferase (ChAT). Combining this histological approach with real time quantification of mRNA levels of nicotinic acetylcholine receptors, we demonstrated that in the TA of Tg2576 mice, neuronal innervation is significantly reduced and synaptic area is smaller and displays less ChAT content when compared to wild type mice. Our study provides the first evidence of reduced cholinergic innervation of skeletal muscle in a mouse model of Aß accumulation. This evidence sustains the possibility that sarcopenia in AD originates from Aß-mediated cholinergic loss.

15.
Curr Alzheimer Res ; 17(3): 259-268, 2020.
Article in English | MEDLINE | ID: mdl-32091333

ABSTRACT

BACKGROUND: Clinical evidence indicates that patients affected by Alzheimer's Disease (AD) fail to form new memories although their memories for old events are intact. This amnesic pattern depends on the selective vulnerability to AD-neurodegeneration of the hippocampus, the brain region that sustains the formation of new memories, while cortical regions that store remote memories are spared. OBJECTIVE: To identify the cellular mechanisms underlying impaired recent memories and intact remote memories in a mouse model of AD. METHODS: Glutamatergic synaptic currents were recorded by patch-clamp in acute hippocampal and anterior Cingulate Cortical (aCC) slices of AD-like Tg2576 mice and Wild-type (Wt) littermates subjected to the Contextual Fear Conditioning (CFC) task or in naïve conditions. RESULTS: We identified a deficit in the formation of recent memories, but not in the recall of remote ones, in Tg2576 mice. With electrophysiological recordings, we detected CFC-induced modifications of the AMPA/NMDA ratio in CA1 pyramidal cells of Wt, but not Tg2576, mice one day after training. CFC-induced changes in the AMPA/NMDA ratio were also detected in the aCC of both Wt and Tg2576 mice 8 days after training. CONCLUSION: Our data suggest that in the early AD stages synaptic plasticity of CA1 synapses, crucial to form new memories, is lost, while plasticity of aCC synapses is intact and contributes to the persistence of long-term memories.


Subject(s)
Alzheimer Disease/physiopathology , Amnesia, Anterograde/physiopathology , CA1 Region, Hippocampal/physiology , Gyrus Cinguli/physiology , Memory, Long-Term/physiology , Neuronal Plasticity/physiology , Amyloid beta-Protein Precursor , Animals , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic , Synaptic Transmission/physiology
16.
Aging Cell ; 19(9): e13189, 2020 09.
Article in English | MEDLINE | ID: mdl-32729663

ABSTRACT

Autophagy agonists have been proposed to slow down neurodegeneration. Spermidine, a polyamine that acts as an autophagy agonist, is currently under clinical trial for the treatment of age-related memory decline. How Spermidine and other autophagy agonists regulate memory and synaptic plasticity is under investigation. We set up a novel mouse model of mild cognitive impairment (MCI), in which middle-aged (12-month-old) mice exhibit impaired memory capacity, lysosomes engulfed with amyloid fibrils (ß-amyloid and α-synuclein) and impaired task-induced GluA1 hippocampal post-translation modifications. Subchronic treatment with Spermidine as well as the autophagy agonist TAT-Beclin 1 rescued memory capacity and GluA1 post-translational modifications by favouring the autophagy/lysosomal-mediated degradation of amyloid fibrils. These findings provide new mechanistic evidence on the therapeutic relevance of autophagy enhancers which, by improving the degradation of misfolded proteins, slow down age-related memory decline.


Subject(s)
Autophagy/genetics , Cognitive Dysfunction/genetics , Memory/drug effects , Aging , Animals , Disease Models, Animal , Mice
17.
iScience ; 23(5): 101078, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32361506

ABSTRACT

Early in brain development, impaired neuronal signaling during time-sensitive windows triggers the onset of neurodevelopmental disorders. GABA, through its depolarizing and excitatory actions, drives early developmental events including neuronal circuit formation and refinement. BDNF/TrkB signaling cooperates with GABA actions. How these developmental processes influence the formation of neural circuits and affect adult brain function is unknown. Here, we show that early deletion of Ntrk2/Trkb from immature mouse hippocampal dentate granule cells (DGCs) affects the integration and maturation of newly formed DGCs in the hippocampal circuitry and drives a premature shift from depolarizing to hyperpolarizing GABAergic actions in the target of DGCs, the CA3 principal cells of the hippocampus, by reducing the expression of the cation-chloride importer Nkcc1. These changes lead to the disruption of early synchronized neuronal activity at the network level and impaired morphological maturation of CA3 pyramidal neurons, ultimately contributing to altered adult hippocampal synaptic plasticity and cognitive processes.

19.
Cell Death Differ ; 27(3): 934-948, 2020 03.
Article in English | MEDLINE | ID: mdl-31591472

ABSTRACT

Alterations of adult neurogenesis have been reported in several Alzheimer's disease (AD) animal models and human brains, while defects in this process at presymptomatic/early stages of AD have not been explored yet. To address this, we investigated potential neurogenesis defects in Tg2576 transgenic mice at 1.5 months of age, a prodromal asymptomatic age in terms of Aß accumulation and neurodegeneration. We observe that Tg2576 resident and SVZ-derived adult neural stem cells (aNSCs) proliferate significantly less. Further, they fail to terminally differentiate into mature neurons due to pathological, tau-mediated, and microtubule hyperstabilization. Olfactory bulb neurogenesis is also strongly reduced, confirming the neurogenic defect in vivo. We find that this phenotype depends on the formation and accumulation of intracellular A-beta oligomers (AßOs) in aNSCs. Indeed, impaired neurogenesis of Tg2576 progenitors is remarkably rescued both in vitro and in vivo by the expression of a conformation-specific anti-AßOs intrabody (scFvA13-KDEL), which selectively interferes with the intracellular generation of AßOs in the endoplasmic reticulum (ER). Altogether, our results demonstrate that SVZ neurogenesis is impaired already at a presymptomatic stage of AD and is caused by endogenously generated intracellular AßOs in the ER of aNSCs. From a translational point of view, impaired SVZ neurogenesis may represent a novel biomarker for AD early diagnosis, in association to other biomarkers. Further, this study validates intracellular Aß oligomers as a promising therapeutic target and prospects anti-AßOs scFvA13-KDEL intrabody as an effective tool for AD treatment.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Intracellular Space/metabolism , Nerve Degeneration/complications , Neurogenesis , Protein Multimerization , Animals , Cell Differentiation , Cell Proliferation , Humans , Mice, Transgenic , Microtubules/metabolism , Neural Stem Cells/metabolism , Neurons/pathology , Olfactory Bulb/metabolism , Protein Conformation
20.
Eur J Neurosci ; 29(5): 902-10, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19291221

ABSTRACT

The phosphodiesterase type IV inhibitor rolipram increases cAMP response element-binding protein (CREB) phosphorylation and exerts neuroprotective effects in both the quinolinic acid rat model of Huntington's disease (DeMarch et al., 2007) and the R6/2 mouse including sparing of striatal neurons, prevention of neuronal intranuclear inclusion formation and attenuation of microglial reaction (DeMarch et al., 2008). In this study, we sought to determine if rolipram has a beneficial role in the altered distribution of CREB binding protein in striatal spiny neurons and in the motor impairments shown by R6/2 mutants. Moreover, we investigated whether rolipram treatment altered the degeneration of parvalbuminergic interneurons typical of Huntington's disease (Fusco et al., 1999). Transgenic mice and their wild-type controls from a stable colony maintained in our laboratory were treated with rolipram (1.5 mg/kg) or saline daily starting from 4 weeks of age. The cellular distribution of CREB binding protein in striatal spiny neurons was assessed by immunofluorescence, whereas parvalbuminergic neuron degeneration was evaluated by cell counts of immunohistochemically labeled tissue. Motor coordination and motor activity were also examined. We found that rolipram was effective in preventing CREB binding protein sequestration into striatal neuronal intranuclear inclusions, sparing parvalbuminergic interneurons of R6/2 mice, and rescuing their motor coordination and motor activity deficits. Our findings demonstrate the possibility of reversing pharmacologically the behavioral and neuropathological abnormalities of symptomatic R6/2 mice and underline the potential therapeutic value of phosphodiesterase type IV inhibitors in Huntington's disease.


Subject(s)
CREB-Binding Protein/metabolism , Corpus Striatum/pathology , Interneurons/metabolism , Movement Disorders/drug therapy , Parvalbumins/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Rolipram/therapeutic use , Analysis of Variance , Animals , Disease Models, Animal , Exploratory Behavior/drug effects , Female , Huntington Disease/complications , Huntington Disease/genetics , Interneurons/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Movement Disorders/etiology , Movement Disorders/pathology , Phosphodiesterase Inhibitors/pharmacology , Protein Transport/drug effects , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Rolipram/pharmacology , Trinucleotide Repeats/genetics , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL