Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Neuron ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38810652

ABSTRACT

Down syndrome (DS) is the most common genetic cause of cognitive disability. However, it is largely unclear how triplication of a small gene subset may impinge on diverse aspects of DS brain physiopathology. Here, we took a multi-omic approach and simultaneously analyzed by RNA-seq and proteomics the expression signatures of two diverse regions of human postmortem DS brains. We found that the overexpression of triplicated genes triggered global expression dysregulation, differentially affecting transcripts, miRNAs, and proteins involved in both known and novel biological candidate pathways. Among the latter, we observed an alteration in RNA splicing, specifically modulating the expression of genes involved in cytoskeleton and axonal dynamics in DS brains. Accordingly, we found an alteration in axonal polarization in neurons from DS human iPSCs and mice. Thus, our study provides an integrated multilayer expression database capable of identifying new potential targets to aid in designing future clinical interventions for DS.

2.
iScience ; 27(4): 109438, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38544574

ABSTRACT

Gamma-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in adults. Depolarizing GABA responses have been well characterized at neuronal-population average level during typical neurodevelopment and partially in brain disorders. However, no investigation has specifically assessed whether a mosaicism of cells with either depolarizing or hyperpolarizing/inhibitory GABAergic responses exists in animals in health/disease at diverse developmental stages, including adulthood. Here, we showed that such mosaicism is present in wild-type (WT) and down syndrome (DS) neuronal networks, as assessed at increasing scales of complexity (cultures, brain slices, behaving mice). Nevertheless, WT mice presented a much lower percentage of cells with depolarizing GABA than DS mice. Restoring the mosaicism of hyperpolarizing and depolarizing GABA-responding neurons to WT levels rescued anxiety behavior in DS mice. Moreover, we found heterogeneous GABAergic responses in developed control and trisomic human induced-pluripotent-stem-cells-derived neurons. Thus, a heterogeneous subpopulation of GABA-responding cells exists in physiological/pathological conditions in mouse and human neurons, possibly contributing to disease-associated behaviors.

3.
Nat Commun ; 14(1): 603, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36746939

ABSTRACT

Spinobulbar muscular atrophy (SBMA) is caused by CAG expansions in the androgen receptor gene. Androgen binding to polyQ-expanded androgen receptor triggers SBMA through a combination of toxic gain-of-function and loss-of-function mechanisms. Leveraging cell lines, mice, and patient-derived specimens, we show that androgen receptor co-regulators lysine-specific demethylase 1 (LSD1) and protein arginine methyltransferase 6 (PRMT6) are overexpressed in an androgen-dependent manner specifically in the skeletal muscle of SBMA patients and mice. LSD1 and PRMT6 cooperatively and synergistically transactivate androgen receptor, and their effect is enhanced by expanded polyQ. Pharmacological and genetic silencing of LSD1 and PRMT6 attenuates polyQ-expanded androgen receptor transactivation in SBMA cells and suppresses toxicity in SBMA flies, and a preclinical approach based on miRNA-mediated silencing of LSD1 and PRMT6 attenuates disease manifestations in SBMA mice. These observations suggest that targeting overexpressed co-regulators can attenuate androgen receptor toxic gain-of-function without exacerbating loss-of-function, highlighting a potential therapeutic strategy for patients with SBMA.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked , Diptera , Muscular Disorders, Atrophic , Mice , Animals , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Bulbo-Spinal Atrophy, X-Linked/genetics , Androgens , Gain of Function Mutation , Phenotype , Histone Demethylases/genetics , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism
4.
J Med Chem ; 64(14): 10203-10229, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34137257

ABSTRACT

Intracellular chloride concentration [Cl-]i is defective in several neurological disorders. In neurons, [Cl-]i is mainly regulated by the action of the Na+-K+-Cl- importer NKCC1 and the K+-Cl- exporter KCC2. Recently, we have reported the discovery of ARN23746 as the lead candidate of a novel class of selective inhibitors of NKCC1. Importantly, ARN23746 is able to rescue core symptoms of Down syndrome (DS) and autism in mouse models. Here, we describe the discovery and extensive characterization of this chemical class of selective NKCC1 inhibitors, with focus on ARN23746 and other promising derivatives. In particular, we present compound 40 (ARN24092) as a backup/follow-up lead with in vivo efficacy in a mouse model of DS. These results further strengthen the potential of this new class of compounds for the treatment of core symptoms of brain disorders characterized by the defective NKCC1/KCC2 expression ratio.


Subject(s)
Down Syndrome/drug therapy , Drug Design , Solute Carrier Family 12, Member 2/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Down Syndrome/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Structure , Structure-Activity Relationship
5.
Mol Ther ; 29(10): 3072-3092, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34058387

ABSTRACT

A common feature of diverse brain disorders is the alteration of GABA-mediated inhibition because of aberrant, intracellular chloride homeostasis induced by changes in the expression and/or function of chloride transporters. Notably, pharmacological inhibition of the chloride importer NKCC1 is able to rescue brain-related core deficits in animal models of these pathologies and in some human clinical studies. Here, we show that reducing NKCC1 expression by RNA interference in the Ts65Dn mouse model of Down syndrome (DS) restores intracellular chloride concentration, efficacy of gamma-aminobutyric acid (GABA)-mediated inhibition, and neuronal network dynamics in vitro and ex vivo. Importantly, adeno-associated virus (AAV)-mediated, neuron-specific NKCC1 knockdown in vivo rescues cognitive deficits in diverse behavioral tasks in Ts65Dn animals. Our results highlight a mechanistic link between NKCC1 expression and behavioral abnormalities in DS mice and establish a molecular target for new therapeutic approaches, including gene therapy, to treat brain disorders characterized by neuronal chloride imbalance.


Subject(s)
Down Syndrome/therapy , Genetic Therapy/methods , Solute Carrier Family 12, Member 2/genetics , Animals , Chlorides/metabolism , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/psychology , Gene Knockdown Techniques , Homeostasis , Male , Mice , Neurons/metabolism , RNA Interference
6.
Neuron ; 108(5): 887-904.e12, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33027640

ABSTRACT

Microglia are brain-resident immune cells and regulate mechanisms essential for cognitive functions. Down syndrome (DS), the most frequent cause of genetic intellectual disability, is caused by a supernumerary chromosome 21, containing also genes related to the immune system. In the hippocampus of the Dp(16) mouse model of DS and DS individuals, we found activated microglia, as assessed by their morphology; activation markers; and, for DS mice, electrophysiological profile. Accordingly, we found increased pro-inflammatory cytokine levels and altered interferon signaling in Dp(16) hippocampi. DS mice also showed decreased spine density and activity of hippocampal neurons and hippocampus-dependent cognitive behavioral deficits. Depletion of defective microglia or treatment with a commonly used anti-inflammatory drug rescued the neuronal spine and activity impairments and cognitive deficits in juvenile Dp(16) mice. Our results suggest an involvement of microglia in Dp(16)-mouse cognitive deficits and identify a new potential therapeutic approach for cognitive disabilities in DS individuals.


Subject(s)
Cognition/physiology , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/physiopathology , Microglia/physiology , Adult , Age Factors , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cognition/drug effects , Down Syndrome/drug therapy , Female , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Pyrroles/pharmacology , Pyrroles/therapeutic use
7.
Chem ; 6(8): 2073-2096, 2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32818158

ABSTRACT

Aberrant expression ratio of Cl- transporters, NKCC1 and KCC2, is implicated in several brain conditions. NKCC1 inhibition by the FDA-approved diuretic drug, bumetanide, rescues core symptoms in rodent models and/or clinical trials with patients. However, bumetanide has a strong diuretic effect due to inhibition of the kidney Cl- transporter NKCC2, creating critical drug compliance issues and health concerns. Here, we report the discovery of a new chemical class of selective NKCC1 inhibitors and the lead drug candidate ARN23746. ARN23746 restores the physiological intracellular Cl- in murine Down syndrome neuronal cultures, has excellent solubility and metabolic stability, and displays no issues with off-target activity in vitro. ARN23746 recovers core symptoms in mouse models of Down syndrome and autism, with no diuretic effect, nor overt toxicity upon chronic treatment in adulthood. ARN23746 is ready for advanced preclinical/manufacturing studies toward the first sustainable therapeutics for the neurological conditions characterized by impaired Cl- homeostasis.

8.
Mol Ther ; 28(2): 642-652, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31495777

ABSTRACT

Glial cell-derived neurotrophic factor (GDNF) has a potent action in promoting the survival of dopamine (DA) neurons. Several studies indicate that increasing GDNF levels may be beneficial for the treatment of Parkinson's disease (PD) by reducing neurodegeneration of DA neurons. Despite a plethora of preclinical studies showing GDNF efficacy in PD animal models, its application in humans remains questionable for its poor efficacy and side effects due to its uncontrolled, ectopic expression. Here we took advantage of SINEUPs, a new class of antisense long non-coding RNA, that promote translation of partially overlapping sense protein-coding mRNAs with no effects on their mRNA levels. By synthesizing a SINEUP targeting Gdnf mRNA, we were able to increase endogenous GDNF protein levels by about 2-fold. Adeno-associated virus (AAV)9-mediated delivery in the striatum of wild-type (WT) mice led to an increase of endogenous GDNF protein for at least 6 months and the potentiation of the DA system's functions while showing no side effects. Furthermore, SINEUP-GDNF was able to ameliorate motor deficits and neurodegeneration of DA neurons in a PD neurochemical mouse model. Our data indicate that SINEUP-GDNF could represent a new strategy to increase endogenous GDNF protein levels in a more physiological manner for therapeutic treatments of PD.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/genetics , Motor Neurons/metabolism , Parkinson Disease/genetics , RNA Interference , RNA, Untranslated/genetics , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dependovirus/genetics , Disease Models, Animal , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Immunohistochemistry , Mice , Motor Neurons/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phenotype
9.
Front Neurosci ; 13: 945, 2019.
Article in English | MEDLINE | ID: mdl-31551698

ABSTRACT

Inherited retinal dystrophies (IRDs) are a large and heterogeneous group of degenerative diseases caused by mutations in various genes. Given the favorable anatomical and immunological characteristics of the eye, gene therapy holds great potential for their treatment. Our goal is to validate the preservation of visual functions by viral-free homology directed repair (HDR) in an autosomal recessive loss of function mutation. We used a tailored gene editing system based on clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) to prevent retinal photoreceptor death in the retinal degeneration 10 (Rd10) mouse model of retinitis pigmentosa. We tested the gene editing tool in vitro and then used in vivo subretinal electroporation to deliver it to one of the retinas of mouse pups at different stages of photoreceptor differentiation. Three months after gene editing, the treated eye exhibited a higher visual acuity compared to the untreated eye. Moreover, we observed preservation of light-evoked responses both in explanted retinas and in the visual cortex of treated animals. Our study validates a CRISPR/Cas9-based therapy as a valuable new approach for the treatment of retinitis pigmentosa caused by autosomal recessive loss-of-function point mutations.

10.
Nat Methods ; 15(11): 969-976, 2018 11.
Article in English | MEDLINE | ID: mdl-30377377

ABSTRACT

Currently available inhibitory optogenetic tools provide short and transient silencing of neurons, but they cannot provide long-lasting inhibition because of the requirement for high light intensities. Here we present an optimized blue-light-sensitive synthetic potassium channel, BLINK2, which showed good expression in neurons in three species. The channel is activated by illumination with low doses of blue light, and in our experiments it remained active over (tens of) minutes in the dark after the illumination was stopped. This activation caused long periods of inhibition of neuronal firing in ex vivo recordings of mouse neurons and impaired motor neuron response in zebrafish in vivo. As a proof-of-concept application, we demonstrated that in a freely moving rat model of neuropathic pain, the activation of a small number of BLINK2 channels caused a long-lasting (>30 min) reduction in pain sensation.


Subject(s)
Action Potentials , Hyperalgesia/physiopathology , Neurons/physiology , Optogenetics , Pain/physiopathology , Peripheral Nervous System Diseases/physiopathology , Recombinant Fusion Proteins/metabolism , Animals , Female , Light , Male , Mice, Inbred C57BL , Neurons/cytology , Paclitaxel/toxicity , Pain/chemically induced , Peripheral Nervous System Diseases/chemically induced , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Zebrafish
11.
Oncotarget ; 9(27): 18760-18774, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29721159

ABSTRACT

Adult neurogenesis is emerging as an important player in brain functions and homeostasis, while impaired or altered adult neurogenesis has been associated with a number of neuropsychiatric diseases, such as depression and epilepsy. Here we investigated the possibility that synapsins (Syns) I and II, beyond their known functions in developing and mature neurons, also play a role in adult neurogenesis. We performed a systematic evaluation of the distinct stages of neurogenesis in the hippocampal dentate gyrus of Syn I and Syn II knockout (KO) mice, before (2-months-old) and after (6-months-old) the appearance of the epileptic phenotype. We found that Syns I and II play an important role in the regulation of adult neurogenesis. In juvenile mice, Syn II deletion was associated with a specific decrease in the proliferation of neuronal progenitors, whereas Syn I deletion impaired the survival of newborn neurons. These defects were reverted after the appearance of the epileptic phenotype, with Syn I KO and Syn II KO mice exhibiting significant increases in survival and proliferation, respectively. Interestingly, long-term potentiation dependent on newborn neurons was present in both juvenile Syn mutants while, at later ages, it was only preserved in Syn II KO mice that also displayed an increased expression of brain-derived neurotrophic factor. This study suggests that Syns I and II play a role in adult neurogenesis and the defects in neurogenesis associated with Syn deletion may contribute to the alterations of cognitive functions observed in Syn-deficient mice.

12.
J Cell Sci ; 131(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29361543

ABSTRACT

Development of remote stimulation techniques for neuronal tissues represents a challenging goal. Among the potential methods, mechanical stimuli are the most promising vectors to convey information non-invasively into intact brain tissue. In this context, selective mechano-sensitization of neuronal circuits would pave the way to develop a new cell-type-specific stimulation approach. We report here, for the first time, the development and characterization of mechano-sensitized neuronal networks through the heterologous expression of an engineered bacterial large-conductance mechanosensitive ion channel (MscL). The neuronal functional expression of the MscL was validated through patch-clamp recordings upon application of calibrated suction pressures. Moreover, we verified the effective development of in-vitro neuronal networks expressing the engineered MscL in terms of cell survival, number of synaptic puncta and spontaneous network activity. The pure mechanosensitivity of the engineered MscL, with its wide genetic modification library, may represent a versatile tool to further develop a mechano-genetic approach.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Escherichia coli Proteins/genetics , Ion Channels/genetics , Mechanotransduction, Cellular/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , Animals , Brain/growth & development , Brain/metabolism , Cell Survival/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/genetics , Ion Channel Gating/genetics , Nerve Net/growth & development , Nerve Net/metabolism , Patch-Clamp Techniques , Primary Cell Culture , Protein Engineering/methods , Rats , Transfection
13.
Sci Rep ; 7(1): 16825, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203796

ABSTRACT

Down syndrome (DS) is caused by the triplication of human chromosome 21 and represents the most frequent genetic cause of intellectual disability. The trisomic Ts65Dn mouse model of DS shows synaptic deficits and reproduces the essential cognitive disabilities of the human syndrome. Aerobic exercise improved various neurophysiological dysfunctions in Ts65Dn mice, including hippocampal synaptic deficits, by promoting synaptogenesis and neurotransmission at glutamatergic terminals. Most importantly, the same intervention also prompted the recovery of hippocampal adult neurogenesis and synaptic plasticity and restored cognitive performance in trisomic mice. Additionally, the expression of brain-derived neurotrophic factor (BDNF) was markedly decreased in the hippocampus of patients with DS. Since the positive effect of exercise was paralleled by increased BDNF expression in trisomic mice, we investigated the effectiveness of a BDNF-mimetic treatment with 7,8-dihydroxyflavone at alleviating intellectual disabilities in the DS model. Pharmacological stimulation of BDNF signaling rescued synaptic plasticity and memory deficits in Ts65Dn mice. Based on our findings, Ts65Dn mice benefit from interventions aimed at promoting brain plasticity, and we provide evidence that BDNF signaling represents a potentially new pharmacological target for treatments aimed at rescuing cognitive disabilities in patients with DS.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Down Syndrome/pathology , Flavones/pharmacology , Learning/drug effects , Memory/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Down Syndrome/drug therapy , Excitatory Postsynaptic Potentials/drug effects , Female , Flavones/therapeutic use , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis , Neuronal Plasticity/drug effects , Physical Conditioning, Animal , Signal Transduction/drug effects
14.
Sci Rep ; 7(1): 4897, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28687747

ABSTRACT

Lamin B1, a key component of the nuclear lamina, plays an important role in brain development. Ablation of endogenous Lamin B1 (Lmnb1) in the mouse strongly impairs embryonic brain development and corticogenesis. However, the mechanisms underlying these neurodevelopmental effects are unknown. Here, we report that Lamin B1 levels modulate the differentiation of murine neural stem cells (NSCs) into neurons and astroglial-like cells. In vitro, endogenous Lmnb1 depletion favors NSC differentiation into glial fibrillar acidic protein (GFAP)-immunoreactive cells over neurons, while overexpression of human Lamin B1 (LMNB1) increases the proportion of neurons. In Lmnb1-null embryos, neurogenesis is reduced, while in vivo Lmnb1 silencing in mouse embryonic brain by in utero electroporation of a specific Lmnb1 sh-RNA results in aberrant cortical positioning of neurons and increased expression of the astrocytic marker GFAP in the cortex of 7-day old pups. Together, these results indicate that finely tuned levels of Lamin B1 are required for NSC differentiation into neurons, proper expression of the astrocytic marker GFAP and corticogenesis.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Glial Fibrillary Acidic Protein/genetics , Lamin Type B/genetics , Neurogenesis/genetics , Neurons/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Cell Differentiation , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/metabolism , Lamin Type B/antagonists & inhibitors , Lamin Type B/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
15.
Cell Rep ; 20(2): 333-343, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28700936

ABSTRACT

Alternative splicing of pre-mRNAs is prominent in the mammalian brain, where it is thought to expand proteome diversity. For example, alternative splicing of voltage-gated Ca2+ channel (VGCC) α1 subunits can generate thousands of isoforms with differential properties and expression patterns. However, the impact of this molecular diversity on brain function, particularly on synaptic transmission, which crucially depends on VGCCs, is unclear. Here, we investigate how two major splice isoforms of P/Q-type VGCCs (Cav2.1[EFa/b]) regulate presynaptic plasticity in hippocampal neurons. We find that the efficacy of P/Q-type VGCC isoforms in supporting synaptic transmission is markedly different, with Cav2.1[EFa] promoting synaptic depression and Cav2.1[EFb] synaptic facilitation. Following a reduction in network activity, hippocampal neurons upregulate selectively Cav2.1[EFa], the isoform exhibiting the higher synaptic efficacy, thus effectively supporting presynaptic homeostatic plasticity. Therefore, the balance between VGCC splice variants at the synapse is a key factor in controlling neurotransmitter release and presynaptic plasticity.


Subject(s)
Alternative Splicing/physiology , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Alternative Splicing/genetics , Animals , Cells, Cultured , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Antagonists/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Picrotoxin/pharmacology , RNA Interference , Rats , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
16.
Front Cell Neurosci ; 11: 54, 2017.
Article in English | MEDLINE | ID: mdl-28326014

ABSTRACT

Down syndrome (DS) is a genetic disorder caused by the presence of a third copy of chromosome 21. DS affects multiple organs, but it invariably results in altered brain development and diverse degrees of intellectual disability. A large body of evidence has shown that synaptic deficits and memory impairment are largely determined by altered GABAergic signaling in trisomic mouse models of DS. These alterations arise during brain development while extending into adulthood, and include genesis of GABAergic neurons, variation of the inhibitory drive and modifications in the control of neural-network excitability. Accordingly, different pharmacological interventions targeting GABAergic signaling have proven promising preclinical approaches to rescue cognitive impairment in DS mouse models. In this review, we will discuss recent data regarding the complex scenario of GABAergic dysfunctions in the trisomic brain of DS mice and patients, and we will evaluate the state of current clinical research targeting GABAergic signaling in individuals with DS.

17.
Sci Transl Med ; 8(370): 370ra181, 2016 12 21.
Article in English | MEDLINE | ID: mdl-28003546

ABSTRACT

Spinobulbar muscular atrophy (SBMA) is an X-linked neuromuscular disease caused by polyglutamine (polyQ) expansion in the androgen receptor (AR) gene. SBMA belongs to the family of polyQ diseases, which are fatal neurodegenerative disorders mainly caused by protein-mediated toxic gain-of-function mechanisms and characterized by deposition of misfolded proteins in the form of aggregates. The neurotoxicity of the polyQ proteins can be modified by phosphorylation at specific sites, thereby providing the rationale for the development of disease-specific treatments. We sought to identify signaling pathways that modulate polyQ-AR phosphorylation for therapy development. We report that cyclin-dependent kinase 2 (CDK2) phosphorylates polyQ-AR specifically at Ser96 Phosphorylation of polyQ-AR by CDK2 increased protein stabilization and toxicity and is negatively regulated by the adenylyl cyclase (AC)/protein kinase A (PKA) signaling pathway. To translate these findings into therapy, we developed an analog of pituitary adenylyl cyclase activating polypeptide (PACAP), a potent activator of the AC/PKA pathway. Chronic intranasal administration of the PACAP analog to knock-in SBMA mice reduced Ser96 phosphorylation, promoted polyQ-AR degradation, and ameliorated disease outcome. These results provide proof of principle that noninvasive therapy based on the use of PACAP analogs is a therapeutic option for SBMA.


Subject(s)
Muscular Disorders, Atrophic/metabolism , Peptides/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Androgen/metabolism , Animals , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase 2/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , Ligands , Membrane Potential, Mitochondrial , Mice , Mice, Transgenic , PC12 Cells , Phosphorylation , Protein Denaturation , Protein Folding , Rats , Rats, Sprague-Dawley , Signal Transduction
18.
J Cell Sci ; 129(9): 1878-91, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26985064

ABSTRACT

L1 (also known as L1CAM) is a trans-membrane glycoprotein mediating neuron-neuron adhesion through homophilic and heterophilic interactions. Although experimental evidence has implicated L1 in axonal outgrowth, fasciculation and pathfinding, its contribution to voltage-gated Na(+) channel function and membrane excitability has remained unknown. Here, we show that firing rate, single cell spiking frequency and Na(+) current density are all reduced in hippocampal excitatory neurons from L1-deficient mice both in culture and in slices owing to an overall reduced membrane expression of Na(+) channels. Remarkably, normal firing activity was restored when L1 was reintroduced into L1-deficient excitatory neurons, indicating that abnormal firing patterns are not related to developmental abnormalities, but are a direct consequence of L1 deletion. Moreover, L1 deficiency leads to impairment of action potential initiation, most likely due to the loss of the interaction of L1 with ankyrin G that produces the delocalization of Na(+) channels at the axonal initial segment. We conclude that L1 contributes to functional expression and localization of Na(+) channels to the neuronal plasma membrane, ensuring correct initiation of action potential and normal firing activity.


Subject(s)
Cell Membrane/metabolism , Gene Expression Regulation/physiology , Hippocampus/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurons/metabolism , Voltage-Gated Sodium Channels/biosynthesis , Animals , Cell Membrane/genetics , Hippocampus/cytology , Mice , Mice, Knockout , Neural Cell Adhesion Molecule L1/genetics , Neurons/cytology , Voltage-Gated Sodium Channels/genetics
19.
Biochim Biophys Acta ; 1862(6): 1093-104, 2016 06.
Article in English | MEDLINE | ID: mdl-26964795

ABSTRACT

Mitochondrial dysfunctions critically impair nervous system development and are potentially involved in the pathogenesis of various neurodevelopmental disorders, including Down syndrome (DS), the most common genetic cause of intellectual disability. Previous studies from our group demonstrated impaired mitochondrial activity in peripheral cells from DS subjects and the efficacy of epigallocatechin-3-gallate (EGCG) - a natural polyphenol major component of green tea - to counteract the mitochondrial energy deficit. In this study, to gain insight into the possible role of mitochondria in DS intellectual disability, mitochondrial functions were analyzed in neural progenitor cells (NPCs) isolated from the hippocampus of Ts65Dn mice, a widely used model of DS which recapitulates many major brain structural and functional phenotypes of the syndrome, including impaired hippocampal neurogenesis. We found that, during NPC proliferation, mitochondrial bioenergetics and mitochondrial biogenic program were strongly compromised in Ts65Dn cells, but not associated with free radical accumulation. These data point to a central role of mitochondrial dysfunction as an inherent feature of DS and not as a consequence of cell oxidative stress. Further, we disclose that, besides EGCG, also the natural polyphenol resveratrol, which displays a neuroprotective action in various human diseases but never tested in DS, restores oxidative phosphorylation efficiency and mitochondrial biogenesis, and improves proliferation of NPCs. These effects were associated with the activation of PGC-1α/Sirt1/AMPK axis by both polyphenols. This research paves the way for using nutraceuticals as a potential therapeutic tool in preventing or managing some energy deficit-associated DS clinical manifestations.


Subject(s)
Antioxidants/therapeutic use , Catechin/analogs & derivatives , Down Syndrome/drug therapy , Hippocampus/drug effects , Mitochondria/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Resveratrol/therapeutic use , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Animals , Catechin/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Down Syndrome/metabolism , Down Syndrome/physiopathology , Female , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice , Mitochondria/metabolism , Mitochondria/pathology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neuroprotective Agents/therapeutic use , Protein Kinases/metabolism
20.
Nat Med ; 21(4): 318-26, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25774849

ABSTRACT

Down syndrome (DS) is the most frequent genetic cause of intellectual disability, and altered GABAergic transmission through Cl(-)-permeable GABAA receptors (GABAARs) contributes considerably to learning and memory deficits in DS mouse models. However, the efficacy of GABAergic transmission has never been directly assessed in DS. Here GABAAR signaling was found to be excitatory rather than inhibitory, and the reversal potential for GABAAR-driven Cl(-) currents (ECl) was shifted toward more positive potentials in the hippocampi of adult DS mice. Accordingly, hippocampal expression of the cation Cl(-) cotransporter NKCC1 was increased in both trisomic mice and individuals with DS. Notably, NKCC1 inhibition by the FDA-approved drug bumetanide restored ECl, synaptic plasticity and hippocampus-dependent memory in adult DS mice. Our findings demonstrate that GABA is excitatory in adult DS mice and identify a new therapeutic approach for the potential rescue of cognitive disabilities in individuals with DS.


Subject(s)
Down Syndrome/metabolism , Memory/physiology , Neuronal Plasticity , Receptors, GABA-A/metabolism , Adolescent , Adult , Animals , Behavior, Animal , Bumetanide/chemistry , Crosses, Genetic , Disease Models, Animal , Female , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Patch-Clamp Techniques , Signal Transduction , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...