Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
EMBO Rep ; 24(8): e56233, 2023 08 03.
Article En | MEDLINE | ID: mdl-37382163

Cortical neurogenesis depends on the balance between self-renewal and differentiation of apical progenitors (APs). Here, we study the epigenetic control of AP's division mode by focusing on the enzymatic activity of the histone methyltransferase DOT1L. Combining lineage tracing with single-cell RNA sequencing of clonally related cells, we show at the cellular level that DOT1L inhibition increases neurogenesis driven by a shift of APs from asymmetric self-renewing to symmetric neurogenic consumptive divisions. At the molecular level, DOT1L activity prevents AP differentiation by promoting transcription of metabolic genes. Mechanistically, DOT1L inhibition reduces activity of an EZH2/PRC2 pathway, converging on increased expression of asparagine synthetase (ASNS), a microcephaly associated gene. Overexpression of ASNS in APs phenocopies DOT1L inhibition, and also increases neuronal differentiation of APs. Our data suggest that DOT1L activity/PRC2 crosstalk controls AP lineage progression by regulating asparagine metabolism.


Aspartate-Ammonia Ligase , Neural Stem Cells , Aspartate-Ammonia Ligase/metabolism , Cell Differentiation/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics
2.
Neurobiol Dis ; 158: 105455, 2021 10.
Article En | MEDLINE | ID: mdl-34358614

Fatal familial insomnia (FFI) is a dominantly inherited prion disease linked to the D178N mutation in the gene encoding the prion protein (PrP). Symptoms, including insomnia, memory loss and motor abnormalities, appear around 50 years of age, leading to death within two years. No treatment is available. A ten-year clinical trial of doxycycline (doxy) is under way in healthy individuals at risk of FFI to test whether presymptomatic doxy prevents or delays the onset of disease. To assess the drug's effect in a tractable disease model, we used Tg(FFI-26) mice, which accumulate aggregated and protease-resistant PrP in their brains and develop a fatal neurological illness highly reminiscent of FFI. Mice were treated daily with 10 mg/kg doxy starting from a presymptomatic stage for twenty weeks. Doxy rescued memory deficits and restored circadian motor rhythmicity in Tg(FFI-26) mice. However, it did not prevent the onset and progression of motor dysfunction, clinical signs and progression to terminal disease. Doxy did not change the amount of aggregated and protease-resistant PrP, but reduced microglial activation in the hippocampus. Presymptomatic doxy treatment rescues cognitive impairment and the motor correlates of sleep dysfunction in Tg(FFI-26) mice but does not prevent fatal disease.


Circadian Rhythm/drug effects , Doxycycline/pharmacology , Doxycycline/therapeutic use , Insomnia, Fatal Familial/drug therapy , Memory/drug effects , Recognition, Psychology/drug effects , Animals , Brain/pathology , Disease Progression , Insomnia, Fatal Familial/genetics , Insomnia, Fatal Familial/pathology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Psychomotor Performance/drug effects
3.
Neuroimage ; 237: 118189, 2021 08 15.
Article En | MEDLINE | ID: mdl-34022383

Large scale neuroimaging datasets present the possibility of providing normative distributions for a wide variety of neuroimaging markers, which would vastly improve the clinical utility of these measures. However, a major challenge is our current poor ability to integrate measures across different large-scale datasets, due to inconsistencies in imaging and non-imaging measures across the different protocols and populations. Here we explore the harmonisation of white matter hyperintensity (WMH) measures across two major studies of healthy elderly populations, the Whitehall II imaging sub-study and the UK Biobank. We identify pre-processing strategies that maximise the consistency across datasets and utilise multivariate regression to characterise study sample differences contributing to differences in WMH variations across studies. We also present a parser to harmonise WMH-relevant non-imaging variables across the two datasets. We show that we can provide highly calibrated WMH measures from these datasets with: (1) the inclusion of a number of specific standardised processing steps; and (2) appropriate modelling of sample differences through the alignment of demographic, cognitive and physiological variables. These results open up a wide range of applications for the study of WMHs and other neuroimaging markers across extensive databases of clinical data.


Aging , Biomedical Research , Datasets as Topic , Leukoaraiosis , Multicenter Studies as Topic , Neuroimaging , Adult , Aged , Aged, 80 and over , Biological Specimen Banks , Female , Humans , Leukoaraiosis/diagnostic imaging , Longitudinal Studies , Male , Middle Aged , United Kingdom
4.
Neurobiol Dis ; 153: 105330, 2021 06.
Article En | MEDLINE | ID: mdl-33711491

Traumatic brain injury (TBI) is associated with widespread tau pathology in about 30% of patients surviving late after injury. We previously found that TBI in mice induces the formation of an abnormal form of tau (tauTBI) which progressively spreads from the site of injury to remote brain regions. Intracerebral inoculation of TBI brain homogenates into naïve mice induced progressive tau pathology, synaptic loss and late cognitive decline, suggesting a pivotal role of tauTBI in post-TBI neurodegeneration. However, the possibility that tauTBI was a marker of TBI-associated neurodegeneration rather than a toxic driver of functional decline could not be excluded. Here we employed the nematode C. elegans as a biosensor to test the pathogenic role of TBI generated tau. The motility of this nematode depends on efficient neuromuscular transmission and is exceptionally sensitive to the toxicity of amyloidogenic proteins, providing a tractable model for our tests. We found that worms exposed to brain homogenates from chronic but not acute TBI mice, or from mice in which tauTBI had been transmitted by intracerebral inoculation, had impaired motility and neuromuscular synaptic transmission. Results were similar when worms were given brain homogenates from transgenic mice overexpressing tau P301L, a tauopathy mouse model, suggesting that TBI-induced and mutant tau have similar toxic properties. P301L brain homogenate toxicity was similar in wild-type and ptl-1 knock-out worms, indicating that the nematode tau homolog protein PTL-1 was not required to mediate the toxic effect. Harsh protease digestion to eliminate the protein component of the homogenates, pre-incubation with anti-tau antibodies or tau depletion by immunoprecipitation, abolished the toxicity. Homogenates of chronic TBI brains from tau knock-out mice were not toxic to C. elegans, whereas oligomeric recombinant tau was sufficient to impair their motility. This study indicates that tauTBI impairs motor activity and synaptic transmission in C. elegans and supports a pathogenic role of tauTBI in the long-term consequences of TBI. It also sets the groundwork for the development of a C. elegans-based platform for screening anti-tau compounds.


Brain Injuries, Traumatic/metabolism , Caenorhabditis elegans , Motor Activity/physiology , Neurodegenerative Diseases/metabolism , Neuromuscular Junction/metabolism , tau Proteins/metabolism , Animals , Brain Injuries, Traumatic/physiopathology , Mice , Neurodegenerative Diseases/physiopathology , Neuromuscular Junction/physiopathology , Tauopathies/metabolism , Tauopathies/physiopathology
5.
J Sleep Res ; 30(3): e13187, 2021 06.
Article En | MEDLINE | ID: mdl-32902030

Sleep is severely impaired in patients with Alzheimer's disease. Amyloid-ß deposition in the brain of Alzheimer's disease patients is a key event in its pathogenesis and is associated with disrupted sleep, even before the appearance of cognitive decline. Because soluble amyloid-ß oligomers are the key mediators of synaptic and cognitive dysfunction in Alzheimer's disease and impair long-term memory in rodents, the first aim of this study was to test the hypothesis that amyloid-ß oligomers would directly impair sleep in mice. The cellular prion protein is a cell surface glycoprotein of uncertain function. Because cellular prion protein binds oligomeric amyloid-ß with high affinity and mediates some of its neurotoxic effects, the second aim of the study was to test whether amyloid-ß oligomer-induced sleep alterations were mediated by cellular prion protein. To address these aims, wild-type and cellular prion protein-deficient mice were given acute intracerebroventricular injections (on different days, at lights on) of vehicle and synthetic amyloid-ß oligomers. Compared to vehicle, amyloid-ß oligomers significantly reduced the amount of time spent in non-rapid eye movement sleep by wild-type mice during both the light and dark phases of the light-dark cycle. The amount of time spent in rapid eye movement sleep was reduced during the dark phase. Sleep was also fragmented by amyloid-ß oligomers, as the number of transitions between states increased in post-injection hours 9-24. No such effects were observed in cellular prion protein-deficient mice. These results show that amyloid-ß oligomers do inhibit and fragment sleep, and that these effects are mediated by cellular prion protein.


Amyloid beta-Peptides/metabolism , Prion Proteins/metabolism , Sleep/genetics , Animals , Humans , Mice
7.
PLoS Pathog ; 16(7): e1008654, 2020 07.
Article En | MEDLINE | ID: mdl-32673372

Prion protein (PrP) mutations are linked to genetic prion diseases, a class of phenotypically heterogeneous neurodegenerative disorders with invariably fatal outcome. How mutant PrP triggers neurodegeneration is not known. Synaptic dysfunction precedes neuronal loss but it is not clear whether, and through which mechanisms, disruption of synaptic activity ultimately leads to neuronal death. Here we show that mutant PrP impairs the secretory trafficking of AMPA receptors (AMPARs). Specifically, intracellular retention of the GluA2 subunit results in synaptic exposure of GluA2-lacking, calcium-permeable AMPARs, leading to increased calcium permeability and enhanced sensitivity to excitotoxic cell death. Mutant PrPs linked to different genetic prion diseases affect AMPAR trafficking and function in different ways. Our findings identify AMPARs as pathogenic targets in genetic prion diseases, and support the involvement of excitotoxicity in neurodegeneration. They also suggest a mechanistic explanation for how different mutant PrPs may cause distinct disease phenotypes.


Calcium/metabolism , Neurons/metabolism , PrPSc Proteins/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Cell Death/physiology , Mice , Permeability , Protein Transport/physiology
8.
Neurobiol Dis ; 130: 104498, 2019 10.
Article En | MEDLINE | ID: mdl-31181281

Prion diseases typically involve brain deposition of abnormally folded prion protein, which is associated with activated glia and increased cytokine production. Cyclophilin A (CypA) is a ubiquitous protein with peptidyl prolyl cis-trans isomerase activity, which regulates protein folding, and can be secreted by cells in response to inflammatory stimuli. On the basis of in vitro studies, CypA was proposed to mediate glial activation during prion infection. To investigate the role of CypA in vivo, we inoculated CypA+/+, CypA+/- and CypA-/- mice with the RML prion strain, and recorded the time to onset of neurological signs and to terminal disease, and the astrocyte and microglia response at presymptomatic and symptomatic stages. Time to onset of disease and survival were significantly shorter in CypA-deficient mice than CypA-expressing controls. CypA-deficient mice had significantly greater microglial activation in the presymptomatic stage, and analysis of anti- and pro-inflammatory microglial markers indicated a shift towards a pro-inflammatory phenotype. There was no difference in astrocyte activation. This suggests that CypA contributes to dampening the pro-inflammatory microglial response during the early stage of RML-induced prion disease.


Brain/pathology , Microglia/metabolism , Peptidylprolyl Isomerase/deficiency , Prion Diseases/metabolism , Prion Diseases/pathology , Animals , Brain/metabolism , Mice , Mice, Knockout
9.
Brain ; 141(9): 2685-2699, 2018 09 01.
Article En | MEDLINE | ID: mdl-30084913

Traumatic brain injury is a risk factor for subsequent neurodegenerative disease, including chronic traumatic encephalopathy, a tauopathy mostly associated with repetitive concussion and blast, but not well recognized as a consequence of severe traumatic brain injury. Here we show that a single severe brain trauma is associated with the emergence of widespread hyperphosphorylated tau pathology in a proportion of humans surviving late after injury. In parallel experimental studies, in a model of severe traumatic brain injury in wild-type mice, we found progressive and widespread tau pathology, replicating the findings in humans. Brain homogenates from these mice, when inoculated into the hippocampus and overlying cerebral cortex of naïve mice, induced widespread tau pathology, synaptic loss, and persistent memory deficits. These data provide evidence that experimental brain trauma induces a self-propagating tau pathology, which can be transmitted between mice, and call for future studies aimed at investigating the potential transmissibility of trauma associated tau pathology in humans.


Brain Injuries, Traumatic/complications , Tauopathies/etiology , Tauopathies/physiopathology , Aged , Aged, 80 and over , Animals , Brain/pathology , Brain Concussion/pathology , Brain Injuries, Traumatic/physiopathology , Cerebral Cortex/pathology , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neurodegenerative Diseases/pathology , Phosphorylation , tau Proteins/metabolism , tau Proteins/physiology
10.
Oncotarget ; 8(47): 83038-83051, 2017 Oct 10.
Article En | MEDLINE | ID: mdl-29137322

Recently a range of ocular manifestations such as retinal and lens amyloid-beta accumulation and retinal nerve fiber layer loss have been proposed as potential biomarkers in Alzheimer disease (AD). The TgCRND8 mouse model of AD exhibits age-dependent amyloid ß (Aß) oligomers accumulation and cognitive defects, amyloid plaques and hyperphosphorylated Tau deposition and inflammation. We proved the correlation between ocular pathologies and AD, observing increased levels of p-APP and p-Tau, accumulation of Aß oligomers in the retina, eye, and optic nerve. The accumulation of amyloid markers was significantly stronger in the retinal ganglion cell (RGC) layer, suggesting that RGC might be more susceptible to degeneration. We detected a thinning of the RGC layer as well as RGC death in the retina of TgCRND8 mice, by using a combination of Optical Coherence Tomography (OCT), immunofluorescence, immunohistochemistry and Western blotting techniques. We proved for the first time the key role of C-Jun N-terminal Kinase (JNK) in the ocular degeneration. In support of this, the administration of the JNK inhibitor, D-JNKI1, was able to counteract the Aß and p-Tau accumulation in the retina of TgCRND8 mice, and consequently reduce RGCs loss. These results confirm that degenerative changes in the retina/eye of AD mouse model mirrors the events observed in the brain parenchyma. Ocular changes can be detected by non-invasive imaging techniques, such as OCT, to study and test different therapeutic strategies against degenerative events associated to AD.

11.
J Neurosci ; 37(43): 10278-10289, 2017 10 25.
Article En | MEDLINE | ID: mdl-28924012

Creutzfeldt-Jakob disease (CJD) is a neurodegenerative disorder caused by prion protein (PrP) misfolding, clinically recognized by cognitive and motor deficits, electroencephalographic abnormalities, and seizures. Its neurophysiological bases are not known. To assess the potential involvement of NMDA receptor (NMDAR) dysfunction, we analyzed NMDA-dependent synaptic plasticity in hippocampal slices from Tg(CJD) mice, which model a genetic form of CJD. Because PrP depletion may result in functional upregulation of NMDARs, we also analyzed PrP knock-out (KO) mice. Long-term potentiation (LTP) at the Schaffer collateral-commissural synapses in the CA1 area of ∼100-d-old Tg(CJD) mice was comparable to that of wild-type (WT) controls, but there was an inversion of metaplasticity, with increased GluN2B phosphorylation, which is indicative of enhanced NMDAR activation. Similar but less marked changes were seen in PrP KO mice. At ∼300 d of age, the magnitude of LTP increased in Tg(CJD) mice but decreased in PrP KO mice, indicating divergent changes in hippocampal synaptic responsiveness. Tg(CJD) but not PrP KO mice were intrinsically more susceptible than WT controls to focal hippocampal seizures induced by kainic acid. IL-1ß-positive astrocytes increased in the Tg(CJD) hippocampus, and blocking IL-1 receptor signaling restored normal synaptic responses and reduced seizure susceptibility. These results indicate that alterations in NMDA-dependent glutamatergic transmission in Tg(CJD) mice do not depend solely on PrP functional loss. Moreover, astrocytic IL-1ß plays a role in the enhanced synaptic responsiveness and seizure susceptibility, suggesting that targeting IL-1ß signaling may offer a novel symptomatic treatment for CJD.SIGNIFICANCE STATEMENT Dementia and myoclonic jerks develop in individuals with Creutzfeldt-Jakob disease (CJD), an incurable brain disorder caused by alterations in prion protein structure. These individuals are prone to seizures and have high brain levels of the inflammatory cytokine IL-1ß. Here we show that blocking IL-1ß receptors with anakinra, the human recombinant form of the endogenous IL-1 receptor antagonist used to treat rheumatoid arthritis, normalizes hippocampal neurotransmission and reduces seizure susceptibility in a CJD mouse model. These results link neuroinflammation to defective neurotransmission and the enhanced susceptibility to seizures in CJD and raise the possibility that targeting IL-1ß with clinically available drugs may be beneficial for symptomatic treatment of the disease.


Creutzfeldt-Jakob Syndrome/drug therapy , Disease Models, Animal , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/drug therapy , Animals , Creutzfeldt-Jakob Syndrome/metabolism , Disease Susceptibility , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Random Allocation , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
12.
PLoS Pathog ; 11(4): e1004796, 2015 Apr.
Article En | MEDLINE | ID: mdl-25880443

Fatal familial insomnia (FFI) and a genetic form of Creutzfeldt-Jakob disease (CJD178) are clinically different prion disorders linked to the D178N prion protein (PrP) mutation. The disease phenotype is determined by the 129 M/V polymorphism on the mutant allele, which is thought to influence D178N PrP misfolding, leading to the formation of distinctive prion strains with specific neurotoxic properties. However, the mechanism by which misfolded variants of mutant PrP cause different diseases is not known. We generated transgenic (Tg) mice expressing the mouse PrP homolog of the FFI mutation. These mice synthesize a misfolded form of mutant PrP in their brains and develop a neurological illness with severe sleep disruption, highly reminiscent of FFI and different from that of analogously generated Tg(CJD) mice modeling CJD178. No prion infectivity was detectable in Tg(FFI) and Tg(CJD) brains by bioassay or protein misfolding cyclic amplification, indicating that mutant PrP has disease-encoding properties that do not depend on its ability to propagate its misfolded conformation. Tg(FFI) and Tg(CJD) neurons have different patterns of intracellular PrP accumulation associated with distinct morphological abnormalities of the endoplasmic reticulum and Golgi, suggesting that mutation-specific alterations of secretory transport may contribute to the disease phenotype.


Insomnia, Fatal Familial/genetics , Insomnia, Fatal Familial/physiopathology , Prions/genetics , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Electroencephalography , Magnetic Resonance Imaging , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Mutation , Phenotype , Prion Proteins
13.
Neuron ; 74(2): 300-13, 2012 Apr 26.
Article En | MEDLINE | ID: mdl-22542184

How mutant prion protein (PrP) leads to neurological dysfunction in genetic prion diseases is unknown. Tg(PG14) mice synthesize a misfolded mutant PrP which is partially retained in the neuronal endoplasmic reticulum (ER). As these mice age, they develop ataxia and massive degeneration of cerebellar granule neurons (CGNs). Here, we report that motor behavioral deficits in Tg(PG14) mice emerge before neurodegeneration and are associated with defective glutamate exocytosis from granule neurons due to impaired calcium dynamics. We found that mutant PrP interacts with the voltage-gated calcium channel α(2)δ-1 subunit, which promotes the anterograde trafficking of the channel. Owing to ER retention of mutant PrP, α(2)δ-1 accumulates intracellularly, impairing delivery of the channel complex to the cell surface. Thus, mutant PrP disrupts cerebellar glutamatergic neurotransmission by reducing the number of functional channels in CGNs. These results link intracellular PrP retention to synaptic dysfunction, indicating new modalities of neurotoxicity and potential therapeutic strategies.


Calcium Channels/metabolism , Cerebellum/cytology , Mutation/genetics , Neurons/metabolism , Prions/genetics , Synaptic Transmission/genetics , Age Factors , Animals , Animals, Newborn , Biophysics , Calcium/metabolism , Cells, Cultured , Disease Models, Animal , Electric Stimulation , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Magnetic Resonance Imaging , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Neurons/ultrastructure , Patch-Clamp Techniques , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Diseases/physiopathology , Prion Proteins , Protein Transport/genetics , Reaction Time/genetics , Rotarod Performance Test , Synaptosomes/metabolism
14.
J Biol Chem ; 283(44): 30101-11, 2008 Oct 31.
Article En | MEDLINE | ID: mdl-18701457

Mutations in the human X-linked cyclin-dependent kinase-like 5 (CDKL5) gene have been identified in patients with Rett syndrome (RTT), West syndrome, and X-linked infantile spasms, sharing the common feature of mental retardation and early seizures. CDKL5 is a rather uncharacterized kinase, but its involvement in RTT seems to be explained by the fact that it works upstream of MeCP2, the main cause of Rett syndrome. To understand the role of this kinase for nervous system functions and to address if molecular mechanisms are involved in regulating its distribution and activity, we studied the ontogeny of CDKL5 expression in developing mouse brains by immunostaining and Western blotting. The expression profile of CDKL5 was compared with that of MeCP2. The two proteins share a general expression profile in the adult mouse brain, but CDKL5 levels appear to be highly modulated at the regional level. Its expression is strongly induced in early postnatal stages, and in the adult brain CDKL5 is present in mature neurons, but not in astroglia. Interestingly, the presence of CDKL5 in the cell nucleus varies at the regional level of the adult brain and is developmentally regulated. CDKL5 shuttles between the cytoplasm and the nucleus and the C-terminal tail is involved in localizing the protein to the cytoplasm in a mechanism depending on active nuclear export. Accordingly, Rett derivatives containing disease-causing truncations of the C terminus are constitutively nuclear, suggesting that they might act as gain of function mutations in this cellular compartment.


Gene Expression Regulation, Developmental , Gene Expression Regulation , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Adult , Animals , Brain/metabolism , Cell Nucleus/metabolism , Female , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Protein Structure, Tertiary , Subcellular Fractions/metabolism
15.
J Biol Chem ; 281(42): 32048-56, 2006 Oct 20.
Article En | MEDLINE | ID: mdl-16935860

Mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene have been identified in patients with Rett syndrome, West syndrome, and X-linked infantile spasms sharing the common features of generally intractable early seizures and mental retardation. Disease-causing mutations are distributed in both the catalytic domain and in the large COOH terminus. In this report, we examine the functional consequences of some Rett mutations of CDKL5 together with some synthetically designed derivatives useful to underline the functional domains of the protein. The mutated CDKL5 derivatives have been subjected to in vitro kinase assays and analyzed for phosphorylation of the TEY (Thr-Glu-Tyr) motif within the activation loop, their subcellular localization, and the capacity of CDKL5 to interact with itself. Whereas wild-type CDKL5 autophosphorylates and mediates the phosphorylation of the methyl-CpG-binding protein 2 (MeCP2) in vitro, Rett-mutated proteins show both impaired and increased catalytic activity suggesting that a tight regulation of CDKL5 is required for correct brain functions. Furthermore, we show that CDKL5 can self-associate and mediate the phosphorylation of its own TEY (Thr-Glu-Tyr) motif. Eventually, we show that the COOH terminus regulates CDKL5 properties; in particular, it negatively influences the catalytic activity and is required for its proper sub-nuclear localization. We propose a model in which CDKL5 phosphorylation is required for its entrance into the nucleus whereas a portion of the COOH-terminal domain is responsible for a stable residency in this cellular compartment probably through protein-protein interactions.


Chromosomes, Human, X , Intellectual Disability/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Spasms, Infantile/genetics , Animals , Brain/metabolism , Cell Nucleus/metabolism , Humans , Infant, Newborn , Methyl-CpG-Binding Protein 2/chemistry , Mice , NIH 3T3 Cells , Protein Structure, Tertiary
16.
Hum Mol Genet ; 14(14): 1935-46, 2005 Jul 15.
Article En | MEDLINE | ID: mdl-15917271

Rett syndrome (RTT) is a severe neurodevelopmental disorder almost exclusively affecting females and characterized by a wide spectrum of clinical manifestations. Most patients affected by classic RTT and a smaller percentage of patients with the milder form 'preserved speech variant' have either point mutations or deletions/duplications in the MECP2 gene. Recently, mutations in the CDKL5 gene, coding for a putative kinase, have been found in female patients with a phenotype overlapping with that of RTT. Here, we report two patients with the early seizure variant of RTT, bearing two novel CDKL5 truncating mutations, strengthening the correlation between CDKL5 and RTT. Considering the similar phenotypes caused by mutations in MECP2 and CDKL5, it has been suggested that the two genes play a role in common pathogenic processes. We show here that CDKL5 is a nuclear protein whose expression in the nervous system overlaps with that of MeCP2, during neural maturation and synaptogenesis. Importantly, we demonstrate that MeCP2 and CDKL5 interact both in vivo and in vitro and that CDKL5 is indeed a kinase, which is able to phosphorylate itself and to mediate MeCP2 phosphorylation, suggesting that they belong to the same molecular pathway. Furthermore, this paper contributes to the clarification of the phenotype associated with CDKL5 mutations and indicates that CDKL5 should be analyzed in each patient showing a clinical course similar to RTT but characterized by a lack of an early normal period due to the presence of seizures.


Methyl-CpG-Binding Protein 2/genetics , Protein Serine-Threonine Kinases/genetics , Rett Syndrome/genetics , Seizures/genetics , Age of Onset , Amino Acid Sequence , Animals , Base Sequence , Child , Child, Preschool , DNA Primers , Female , Humans , Immunoprecipitation , In Situ Hybridization , Mice , Molecular Sequence Data , Phosphorylation , Point Mutation , Protein Serine-Threonine Kinases/chemistry , Transcription, Genetic
17.
Neurobiol Aging ; 23(5): 957-76, 2002.
Article En | MEDLINE | ID: mdl-12392798

The accumulation of altered proteins is a common pathogenic mechanism in several neurodegenerative disorders. A causal role of protein aggregation was originally proposed in Alzheimer's disease (AD) where extracellular deposition of beta-amyloid (Abeta) is the main neuropathological feature. It is now believed that intracellular deposition of aggregated proteins may be relevant in Parkinson's disease (PD), amyotrophic lateral sclerosis and polyglutamine disorders. An impairment of ubiquitin-proteasome system (UPS) appears directly involved in these disorders. We reviewed the results on the role of protein misfolding in AD and PD and the influence of mutations associated with these diseases on the expression of amyloidogenic proteins. Results of genetic screening of familial cases of AD and PD are summarized. In the familial AD population (70 subjects) we found several mutations of the presenilin 1 (PS1) gene with a frequency of 12.8% and one mutation in the gene encoding the protein precursor of amyloid (APP) (1.4%). One mutation of Parkin in the homozygous form and two in the heterozygous form were identified in our PD population. We also reported data obtained with synthetic peptides and other experimental models, for evaluation of the pathogenic role of mutations in terms of protein misfolding.


Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases , Aged , Amyloid/chemistry , Amyloid/metabolism , Cysteine Endopeptidases/metabolism , Humans , Ligases/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/metabolism , Presenilin-1 , Presenilin-2 , Proteasome Endopeptidase Complex , Protein Folding , Synucleins , Ubiquitin/metabolism
18.
Proc Natl Acad Sci U S A ; 99(16): 10849-54, 2002 Aug 06.
Article En | MEDLINE | ID: mdl-12149459

Prion diseases are transmissible neurodegenerative disorders of humans and animals for which no effective treatment is available. Conformationally altered, protease-resistant forms of the prion protein (PrP) termed PrP(Sc) are critical for disease transmissibility and pathogenesis, thus representing a primary target for therapeutic strategies. Based on previous findings that tetracyclines revert abnormal physicochemical properties and abolish neurotoxicity of PrP peptides in vitro, we tested the ability of these compounds to interact with PrP(Sc) from patients with the new variant of Creutzfeldt-Jakob disease (vCJD) and cattle with bovine spongiform encephalopathy (BSE). The incubation with tetracycline hydrochloride or doxycycline hyclate at concentrations ranging from 10 microM to 1 mM resulted in a dose-dependent decrease in protease resistance of PrP(Sc). This finding prompted us to investigate whether tetracyclines affect prion infectivity by using an animal model of disease. Syrian hamsters were injected intracerebrally with 263K scrapie-infected brain homogenate that was coincubated with 1 mM tetracycline hydrochloride, 1 mM doxycycline hyclate, or vehicle solution before inoculation. Hamsters injected with tetracycline-treated inoculum showed a significant delay in the onset of clinical signs of disease and prolonged survival time. These effects were paralleled by a delay in the appearance of magnetic-resonance abnormalities in the thalamus, neuropathological changes, and PrP(Sc) accumulation. When tetracycline was preincubated with highly diluted scrapie-infected inoculum, one third of hamsters did not develop disease. Our data suggest that these well characterized antibiotics reduce prion infectivity through a direct interaction with PrP(Sc) and are potentially useful for inactivation of BSE- or vCJD-contaminated products and prevention strategies.


Anti-Bacterial Agents/pharmacology , PrPSc Proteins/drug effects , Tetracycline/pharmacology , Animals , Brain/metabolism , Cattle , Creutzfeldt-Jakob Syndrome/etiology , Cricetinae , Disease Models, Animal , Doxycycline/analogs & derivatives , Doxycycline/pharmacology , Encephalopathy, Bovine Spongiform/etiology , Endopeptidase K/metabolism , Gentamicins/pharmacology , Guanidines/pharmacology , Humans , Isothiocyanates/pharmacology , Mesocricetus , PrPSc Proteins/metabolism , PrPSc Proteins/pathogenicity , Protein Denaturation , Scrapie/etiology
...