Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 31(9): 1389-1406, 2022 05 04.
Article in English | MEDLINE | ID: mdl-34761259

ABSTRACT

Autism spectrum disorder (ASD) and intellectual disability (ID) often exist together in patients. The RAB39B gene has been reported to be mutated in ID patients with additional clinical features ranging from ASD, macrocephaly, seizures and/or early-onset parkinsonism. Here, we describe a novel RAB39B nonstop mutation [Xq28; c.640 T > C; p.(*214Glnext*21)] in a family with ASD, severe ID and poor motor coordination, and we assessed the pathogenicity of the mutation. A heterologous cell system and a Rab39b knockdown (KD) murine model, which mimic the nonstop mutation, were used to validate the deleterious effect of the RAB39B mutation. The mutation led to RAB39B protein instability, resulting in its increased degradation and consequent downregulation. Using a Rab39b KD mouse model, we demonstrated that the downregulation of RAB39B led to increased GluA2 lacking Ca2+-permeable AMPAR composition at the hippocampal neuronal surface and increased dendritic spine density that remained in an immature filopodia-like state. These phenotypes affected behavioural performance in a disease-specific manner. Rab39b KD mice revealed impaired social behaviour but intact social recognition. They also showed normal anxiety-like, exploratory and motivational behaviours but impaired working and associative memories. In conclusion, we found a novel RAB39B nonstop variant that segregated in a family with a clinical phenotype including ID, ASD and poor motor coordination. The pathogenicity of mutations causing the downregulation of RAB39B proteins, impacting AMPAR trafficking and dendritic spine morphogenesis, reinforced the idea that AMPAR modulation and dendritic spine assets could be considered hallmarks of neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Animals , Autism Spectrum Disorder/genetics , Disease Models, Animal , Down-Regulation , Humans , Intellectual Disability/genetics , Mice , Mutation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
2.
Mol Psychiatry ; 26(11): 6531-6549, 2021 11.
Article in English | MEDLINE | ID: mdl-34035473

ABSTRACT

Mutations in the RAB39B gene cause X-linked intellectual disability (XLID), comorbid with autism spectrum disorders or early Parkinson's disease. One of the functions of the neuronal small GTPase RAB39B is to drive GluA2/GluA3 α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) maturation and trafficking, determining AMPAR subunit composition at glutamatergic postsynaptic neuronal terminals. Taking advantage of the Rab39b knockout murine model, we show that a lack of RAB39B affects neuronal dendritic spine refinement, prompting a more Ca2+-permeable and excitable synaptic network, which correlates with an immature spine arrangement and behavioural and cognitive alterations in adult mice. The persistence of immature circuits is triggered by increased hypermobility of the spine, which is restored by the Ca2+-permeable AMPAR antagonist NASPM. Together, these data confirm that RAB39B controls AMPAR trafficking, which in turn plays a pivotal role in neuronal dendritic spine remodelling and that targeting Ca2+-permeable AMPARs may highlight future pharmaceutical interventions for RAB39B-associated disease conditions.


Subject(s)
Dendritic Spines , Intellectual Disability , rab GTP-Binding Proteins , Animals , Calcium , Dendritic Spines/physiology , Mice , Neuronal Plasticity , Neurons/physiology , Receptors, Glutamate/physiology , rab GTP-Binding Proteins/physiology
3.
Hum Mol Genet ; 28(1): 124-132, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30239779

ABSTRACT

Protein zero (P0) is the major structural protein in peripheral myelin, and mutations in the Myelin Protein Zero (Mpz) gene produce wide-ranging hereditary neuropathy phenotypes. To gain insight in the mechanisms underlying a particularly severe form, congenital hypomyelination (CH), we targeted mouse Mpz to encode P0Q215X, a nonsense mutation associated with the disease, that we show escapes nonsense mediated decay and is expressed in CH patient nerves. The knock-in mice express low levels of the resulting truncated protein, producing a milder phenotype when compared to patients, allowing to dissect the subtle pathogenic mechanisms occurring in otherwise very compromised peripheral myelin. We find that P0Q215X does not elicit an unfolded protein response, which is a key mechanism for other pathogenic MPZ mutations, but is instead in part aberrantly trafficked to non-myelin plasma membranes and induces defects in radial sorting of axons by Schwann cells. We show that the loss of the C-terminal Tyr-Ala-Met-Leu motif is responsible for P0 mislocalization, as its addition is able to restore correct P0Q215X trafficking in vitro. Lastly, we show that P0Q215X acts through dose-dependent gain of abnormal function, as wild-type P0 is unable to rescue the hypomyelination phenotype. Collectively, these data indicate that alterations at the premyelinating stage, linked to altered targeting of P0, may be responsible for CH, and that different types of gain of abnormal function produce the diverse neuropathy phenotypes associated with MPZ, supporting future allele-specific therapeutic silencing strategies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Myelin P0 Protein/genetics , Myelin P0 Protein/physiology , Animals , Axons/metabolism , Cell Membrane/physiology , Codon, Nonsense , Demyelinating Diseases/genetics , Female , Gene Knock-In Techniques , Humans , Male , Mice, Inbred BALB C , Mutation , Phenotype , Protein Transport/genetics , Schwann Cells/metabolism
4.
Hum Mol Genet ; 26(23): 4699-4714, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973667

ABSTRACT

Intellectual Disability is a common and heterogeneous disorder characterized by limitations in intellectual functioning and adaptive behaviour, whose molecular mechanisms remain largely unknown. Among the numerous genes found to be involved in the pathogenesis of intellectual disability, 10% are located on the X-chromosome. We identified a missense mutation (c.236 C > G; p.S79W) in the SYN1 gene coding for synapsin I in the MRX50 family, affected by non-syndromic X-linked intellectual disability. Synapsin I is a neuronal phosphoprotein involved in the regulation of neurotransmitter release and neuronal development. Several mutations in SYN1 have been identified in patients affected by epilepsy and/or autism. The S79W mutation segregates with the disease in the MRX50 family and all affected members display intellectual disability as sole clinical manifestation. At the protein level, the S79W Synapsin I mutation is located in the region of the B-domain involved in recognition of highly curved membranes. Expression of human S79W Synapsin I in Syn1 knockout hippocampal neurons causes aberrant accumulation of small clear vesicles in the soma, increased clustering of synaptic vesicles at presynaptic terminals and increased frequency of excitatory spontaneous release events. In addition, the presence of S79W Synapsin I strongly reduces the mobility of synaptic vesicles, with possible implications for the regulation of neurotransmitter release and synaptic plasticity. These results implicate SYN1 in the pathogenesis of non-syndromic intellectual disability, showing that alterations of synaptic vesicle trafficking are one possible cause of this disease, and suggest that distinct mutations in SYN1 may lead to distinct brain pathologies.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation, Missense , Synapsins/genetics , Synaptic Vesicles/genetics , Animals , Base Sequence , Humans , Mental Retardation, X-Linked/metabolism , Mice , Mice, Knockout , Mutation , Neurogenesis/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , Pedigree , Presynaptic Terminals/metabolism , Primary Cell Culture , Protein Transport , Synapsins/metabolism , Synaptic Transmission/genetics , Synaptic Vesicles/metabolism
5.
J Neuroinflammation ; 15(1): 58, 2018 Feb 23.
Article in English | MEDLINE | ID: mdl-29475438

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) is a devastating condition mainly deriving from a traumatic damage of the spinal cord (SC). Immune cells and endogenous SC-neural stem cells (SC-NSCs) play a critical role in wound healing processes, although both are ineffective to completely restore tissue functioning. The role of SC-NSCs in SCI and, in particular, whether such cells can interplay with the immune response are poorly investigated issues, although mechanisms governing such interactions might open new avenues to develop novel therapeutic approaches. METHODS: We used two transgenic mouse lines to trace as well as to kill SC-NSCs in mice receiving SCI. We used Nestin CreERT2 mice to trace SC-NSCs descendants in the spinal cord of mice subjected to SCI. While mice carrying the suicide gene thymidine kinase (TK) along with the GFP reporter, under the control of the Nestin promoter regions (NestinTK mice) were used to label and selectively kill SC-NSCs. RESULTS: We found that SC-NSCs are capable to self-activate after SCI. In addition, a significant worsening of clinical and pathological features of SCI was observed in the NestinTK mice, upon selective ablation of SC-NSCs before the injury induction. Finally, mice lacking in SC-NSCs and receiving SCI displayed reduced levels of different neurotrophic factors in the SC and significantly higher number of M1-like myeloid cells. CONCLUSION: Our data show that SC-NSCs undergo cell proliferation in response to traumatic spinal cord injury. Mice lacking SC-NSCs display overt microglia activation and exaggerate expression of pro-inflammatory cytokines. The absence of SC-NSCs impaired functional recovery as well as neuronal and oligodendrocyte cell survival. Collectively our data indicate that SC-NSCs can interact with microglia/macrophages modulating their activation/responses and that such interaction is importantly involved in mechanisms leading tissue recovery.


Subject(s)
Disease Models, Animal , Locomotion/physiology , Neural Stem Cells/pathology , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord/pathology , Animals , Cell Proliferation/physiology , Male , Mice , Mice, Transgenic , Spinal Cord/physiopathology , Spinal Cord Injuries/physiopathology
6.
Cereb Cortex ; 27(11): 5369-5384, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28968657

ABSTRACT

Intellectual disability affects 2-3% of the world's population and typically begins during childhood, causing impairments in social skills and cognitive abilities. Mutations in the TM4SF2 gene, which encodes the TSPAN7 protein, cause a severe form of intellectual disability, and currently, no therapy is able to ameliorate this cognitive impairment. We previously reported that, in cultured neurons, shRNA-mediated down-regulation of TSPAN7 affects AMPAR trafficking by enhancing PICK1-GluA2 interaction, thereby increasing the intracellular retention of AMPAR. Here, we found that loss of TSPAN7 function in mice causes alterations in hippocampal excitatory synapse structure and functionality as well as cognitive impairment. These changes occurred along with alterations in AMPAR expression levels. We also found that interfering with PICK1-GluA2 binding restored synaptic function in Tm4sf2-/y mice. Moreover, potentiation of AMPAR activity via the administration of the ampakine CX516 reverted the neurological phenotype observed in Tm4sf2-/y mice, suggesting that pharmacological modulation of AMPAR may represent a new approach for treating patients affected by TM4SF2 mutations and intellectual disability.


Subject(s)
Excitatory Amino Acid Agents/pharmacology , Intellectual Disability/drug therapy , Intellectual Disability/metabolism , Membrane Proteins/deficiency , Nerve Tissue Proteins/deficiency , Psychotropic Drugs/pharmacology , Receptors, AMPA/metabolism , Allosteric Regulation , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Disease Models, Animal , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/ultrastructure , Intellectual Disability/pathology , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding/drug effects , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques
7.
Am J Hum Genet ; 95(6): 729-35, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25434005

ABSTRACT

Advances in understanding the etiology of Parkinson disease have been driven by the identification of causative mutations in families. Genetic analysis of an Australian family with three males displaying clinical features of early-onset parkinsonism and intellectual disability identified a ∼45 kb deletion resulting in the complete loss of RAB39B. We subsequently identified a missense mutation (c.503C>A [p.Thr168Lys]) in RAB39B in an unrelated Wisconsin kindred affected by a similar clinical phenotype. In silico and in vitro studies demonstrated that the mutation destabilized the protein, consistent with loss of function. In vitro small-hairpin-RNA-mediated knockdown of Rab39b resulted in a reduction in the density of α-synuclein immunoreactive puncta in dendritic processes of cultured neurons. In addition, in multiple cell models, we demonstrated that knockdown of Rab39b was associated with reduced steady-state levels of α-synuclein. Post mortem studies demonstrated that loss of RAB39B resulted in pathologically confirmed Parkinson disease. There was extensive dopaminergic neuron loss in the substantia nigra and widespread classic Lewy body pathology. Additional pathological features included cortical Lewy bodies, brain iron accumulation, tau immunoreactivity, and axonal spheroids. Overall, we have shown that loss-of-function mutations in RAB39B cause intellectual disability and pathologically confirmed early-onset Parkinson disease. The loss of RAB39B results in dysregulation of α-synuclein homeostasis and a spectrum of neuropathological features that implicate RAB39B in the pathogenesis of Parkinson disease and potentially other neurodegenerative disorders.


Subject(s)
Genes, X-Linked , Intellectual Disability/genetics , Nerve Degeneration/genetics , Parkinson Disease/genetics , alpha-Synuclein/metabolism , rab GTP-Binding Proteins/genetics , Amino Acid Substitution , Australia , Base Sequence , Dopamine/metabolism , Female , Gene Expression Regulation , Humans , Intellectual Disability/physiopathology , Lewy Bodies/metabolism , Male , Middle Aged , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Nerve Degeneration/physiopathology , Parkinson Disease/physiopathology , Pedigree , Sequence Analysis, DNA , Sequence Deletion , Substantia Nigra/physiopathology , rab GTP-Binding Proteins/metabolism
8.
Cereb Cortex ; 26(2): 873-890, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582364

ABSTRACT

Rac GTPases regulate the development of cortical/hippocampal GABAergic interneurons by affecting the early development and migration of GABAergic precursors. We have addressed the function of Rac1 and Rac3 proteins during the late maturation of hippocampal interneurons. We observed specific phenotypic differences between conditional Rac1 and full Rac3 knockout mice. Rac1 deletion caused greater generalized hyperactivity and cognitive impairment compared with Rac3 deletion. This phenotype matched with a more evident functional impairment of the inhibitory circuits in Rac1 mutants, showing higher excitability and reduced spontaneous inhibitory currents in the CA hippocampal pyramidal neurons. Morphological analysis confirmed a differential modification of the inhibitory circuits: deletion of either Rac caused a similar reduction of parvalbumin-positive inhibitory terminals in the pyramidal layer. Intriguingly, cannabinoid receptor-1-positive terminals were strongly increased only in the CA1 of Rac1-depleted mice. This increase may underlie the stronger electrophysiological defects in this mutant. Accordingly, incubation with an antagonist for cannabinoid receptors partially rescued the reduction of spontaneous inhibitory currents in the pyramidal cells of Rac1 mutants. Our results show that Rac1 and Rac3 have independent roles in the formation of GABAergic circuits, as highlighted by the differential effects of their deletion on the late maturation of specific populations of interneurons.


Subject(s)
Behavior, Animal/physiology , GABAergic Neurons/physiology , Hippocampus/cytology , Nerve Net/metabolism , rac GTP-Binding Proteins/deficiency , rac1 GTP-Binding Protein/deficiency , Adaptation, Ocular/genetics , Animals , Conditioning, Classical/physiology , Emotions/physiology , Excitatory Amino Acid Agents/pharmacology , Exploratory Behavior/physiology , Gene Expression Regulation/genetics , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Pyramidal Cells/metabolism , Synapsins/genetics , Synapsins/metabolism , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/genetics
9.
J Am Soc Nephrol ; 27(7): 1958-69, 2016 07.
Article in English | MEDLINE | ID: mdl-26534924

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is an important cause of ESRD for which there exists no approved therapy in the United States. Defective glucose metabolism has been identified as a feature of ADPKD, and inhibition of glycolysis using glucose analogs ameliorates aggressive PKD in preclinical models. Here, we investigated the effects of chronic treatment with low doses of the glucose analog 2-deoxy-d-glucose (2DG) on ADPKD progression in orthologous and slowly progressive murine models created by inducible inactivation of the Pkd1 gene postnatally. As previously reported, early inactivation (postnatal days 11 and 12) of Pkd1 resulted in PKD developing within weeks, whereas late inactivation (postnatal days 25-28) resulted in PKD developing in months. Irrespective of the timing of Pkd1 gene inactivation, cystic kidneys showed enhanced uptake of (13)C-glucose and conversion to (13)C-lactate. Administration of 2DG restored normal renal levels of the phosphorylated forms of AMP-activated protein kinase and its target acetyl-CoA carboxylase. Furthermore, 2DG greatly retarded disease progression in both model systems, reducing the increase in total kidney volume and cystic index and markedly reducing CD45-positive cell infiltration. Notably, chronic administration of low doses (100 mg/kg 5 days per week) of 2DG did not result in any obvious sign of toxicity as assessed by analysis of brain and heart histology as well as behavioral tests. Our data provide proof of principle support for the use of 2DG as a therapeutic strategy in ADPKD.


Subject(s)
Deoxyglucose/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Animals , Disease Models, Animal , Disease Progression , Female , Male , Mice
10.
Brain Behav Immun ; 45: 263-76, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25499583

ABSTRACT

Local acidosis is associated with neuro-inflammation and can have significant effects in several neurological disorders, including multiple sclerosis, brain ischemia, spinal cord injury and epilepsy. Despite local acidosis has been implicated in numerous pathological functions, very little is known about the modulatory effects of pathological acidosis on the activity of neuronal networks and on synaptic structural properties. Using non-invasive MRI spectroscopy we revealed protracted extracellular acidosis in the CNS of Experimental Autoimmune Encephalomyelitis (EAE) affected mice. By multi-unit recording in cortical neurons, we established that acidosis affects network activity, down-sizing firing and bursting behaviors as well as amplitudes. Furthermore, a protracted acidosis reduced the number of presynaptic terminals, while it did not affect the postsynaptic compartment. Application of the diarylamidine Diminazene Aceturate (DA) during acidosis significantly reverted both the loss of neuronal firing and bursting and the reduction of presynaptic terminals. Finally, in vivo DA delivery ameliorated the clinical disease course of EAE mice, reducing demyelination and axonal damage. DA is known to block acid-sensing ion channels (ASICs), which are proton-gated, voltage-insensitive, Na(+) permeable channels principally expressed by peripheral and central nervous system neurons. Our data suggest that ASICs activation during acidosis modulates network electrical activity and exacerbates neuro-degeneration in EAE mice. Therefore pharmacological modulation of ASICs in neuroinflammatory diseases could represent a new promising strategy for future therapies aimed at neuro-protection.


Subject(s)
Acid Sensing Ion Channel Blockers/pharmacology , Acid Sensing Ion Channels/metabolism , Acidosis/metabolism , Brain/metabolism , Diminazene/analogs & derivatives , Encephalomyelitis, Autoimmune, Experimental/metabolism , Myelin Sheath/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Brain/drug effects , Brain/pathology , Diminazene/pharmacology , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Mice , Myelin Sheath/drug effects , Myelin Sheath/pathology , Neurons/drug effects , Neurons/pathology , Presynaptic Terminals/drug effects , Synaptic Potentials/drug effects
11.
Hum Mutat ; 35(3): 377-83, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357492

ABSTRACT

Copy number gains at Xq28 are a frequent cause of X-linked intellectual disability (XLID). Here, we report on a recurrent 0.5 Mb tandem copy number gain at distal Xq28 not including MECP2, in four male patients with nonsyndromic mild ID and behavioral problems. The genomic region is duplicated in two families and triplicated in a third reflected by more distinctive clinical features. The X-inactivation patterns in carrier females correspond well with their clinical symptoms. Our mapping data confirm that this recurrent gain is likely mediated by nonallelic homologous recombination between two directly oriented Int22h repeats. The affected region harbors eight genes of which RAB39B encoding a small GTPase, was the prime candidate since loss-of-function mutations had been linked to ID. RAB39B is expressed at stable levels in lymphocytes from control individuals, suggesting a tight regulation. mRNA levels in our patients were almost two-fold increased. Overexpression of Rab39b in mouse primary hippocampal neurons demonstrated a significant decrease in neuronal branching as well as in the number of synapses when compared with the control neurons. Taken together, we provide evidence that the increased dosage of RAB39B causes a disturbed neuronal development leading to cognitive impairment in patients with this recurrent copy number gain.


Subject(s)
Chromosomes, Human, X/genetics , DNA Copy Number Variations , Intellectual Disability/genetics , rab GTP-Binding Proteins/genetics , Animals , Belgium , Cell Differentiation , Child , Chromosome Mapping , Estonia , Gene Duplication , Gene Expression Regulation , Humans , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Neurons/cytology , Neurons/metabolism , White People , X Chromosome Inactivation
12.
Commun Biol ; 7(1): 140, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38291108

ABSTRACT

Plasma-derived therapeutic proteins are produced through an industrial fractionation process where proteins are purified from individual intermediates, some of which remain unused and are discarded. Relatively few plasma-derived proteins are exploited clinically, with most of available plasma being directed towards the manufacture of immunoglobulin and albumin. Although the plasma proteome provides opportunities to develop novel protein replacement therapies, particularly for rare diseases, the high cost of plasma together with small patient populations impact negatively on the development of plasma-derived orphan drugs. Enabling therapeutics development from unused plasma fractionation intermediates would therefore constitute a substantial innovation. To this objective, we characterized the proteome of unused plasma fractionation intermediates and prioritized proteins for their potential as new candidate therapies for human disease. We selected ceruloplasmin, a plasma ferroxidase, as a potential therapy for aceruloplasminemia, an adult-onset ultra-rare neurological disease caused by iron accumulation as a result of ceruloplasmin mutations. Intraperitoneally administered ceruloplasmin, purified from an unused plasma fractionation intermediate, was able to prevent neurological, hepatic and hematological phenotypes in ceruloplasmin-deficient mice. These data demonstrate the feasibility of transforming industrial waste plasma fraction into a raw material for manufacturing of new candidate proteins for replacement therapies, optimizing plasma use and reducing waste generation.


Subject(s)
Ceruloplasmin , Iron Metabolism Disorders , Neurodegenerative Diseases , Proteome , Adult , Humans , Animals , Mice , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Proteome/metabolism , Rare Diseases , Industrial Waste
13.
Am J Hum Genet ; 86(2): 185-95, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20159109

ABSTRACT

Human Mental Retardation (MR) is a common and highly heterogeneous pediatric disorder affecting around 3% of the general population; at least 215 X-linked MR (XLMR) conditions have been described, and mutations have been identified in 83 different genes, encoding proteins with a variety of function, such as chromatin remodeling, synaptic function, and intracellular trafficking. The small GTPases of the RAB family, which play an essential role in intracellular vesicular trafficking, have been shown to be involved in MR. We report here the identification of mutations in the small GTPase RAB39B gene in two male patients. One mutation in family X (D-23) introduced a stop codon seven amino acids after the start codon (c.21C > A; p.Y7X). A second mutation, in the MRX72 family, altered the 5' splice site (c.215+1G > A) and normal splicing. Neither instance produced a protein. Mutations segregate with the disease in the families, and in some family members intellectual disabilities were associated with autism spectrum disorder, epileptic seizures, and macrocephaly. We show that RAB39B, a novel RAB GTPase of unknown function, is a neuronal-specific protein that is localized to the Golgi compartment. Its downregulation leads to an alteration in the number and morphology of neurite growth cones and a significant reduction in presynaptic buttons, suggesting that RAB39B is required for synapse formation and maintenance. Our results demonstrate developmental and functional neuronal alteration as a consequence of downregulation of RAB39B and emphasize the critical role of vesicular trafficking in the development of neurons and human intellectual abilities.


Subject(s)
Autistic Disorder/complications , Craniofacial Abnormalities/complications , Epilepsy/complications , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/genetics , Mutation/genetics , rab GTP-Binding Proteins/genetics , Animals , Autistic Disorder/genetics , Base Sequence , Brain/metabolism , Brain/pathology , Cell Differentiation , Craniofacial Abnormalities/genetics , DNA Mutational Analysis , Down-Regulation/genetics , Epilepsy/genetics , Female , Golgi Apparatus/metabolism , HeLa Cells , Humans , Male , Mice , Molecular Sequence Data , Neurons/metabolism , Neurons/pathology , Organ Specificity/genetics , Pedigree , Protein Transport , RNA, Small Interfering/metabolism , Synapses/genetics
14.
Am J Pathol ; 180(3): 1121-1135, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22234171

ABSTRACT

Protein synthesis is a tightly regulated, energy-consuming process. The control of mRNA translation into protein is fundamentally important for the fine-tuning of gene expression; additionally, precise translational control plays a critical role in many cellular processes, including development, cellular growth, proliferation, differentiation, synaptic plasticity, memory, and learning. Eukaryotic translation initiation factor 4h (Eif4h) encodes a protein involved in the process of protein synthesis, at the level of initiation phase. Its human homolog, WBSCR1, maps on 7q11.23, inside the 1.6 Mb region that is commonly deleted in patients affected by the Williams-Beuren syndrome, which is a complex neurodevelopmental disorder characterized by cardiovascular defects, cerebral dysplasias and a peculiar cognitive-behavioral profile. In this study, we generated knockout mice deficient in Eif4h. These mice displayed growth retardation with a significant reduction of body weight that began from the first week of postnatal development. Neuroanatomical profiling results generated by magnetic resonance imaging analysis revealed a smaller brain volume in null mice compared with controls as well as altered brain morphology, where anterior and posterior brain regions were differentially affected. The inactivation of Eif4h also led to a reduction in both the number and complexity of neurons. Behavioral studies revealed severe impairments of fear-related associative learning and memory formation. These alterations suggest that Eif4h might contribute to certain deficits associated with Williams-Beuren syndrome.


Subject(s)
Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Growth Disorders/genetics , Learning Disabilities/genetics , Memory Disorders/genetics , Williams Syndrome/genetics , Animals , Brain/growth & development , Brain/metabolism , Conditioning, Psychological/physiology , Eukaryotic Initiation Factors/metabolism , Exploratory Behavior/physiology , Fear , Female , Gene Deletion , Heterozygote , Male , Mice , Mice, Knockout , Muscle Fatigue/genetics , Mutagenesis, Insertional , Organ Size , Psychomotor Performance/physiology , RNA, Messenger/metabolism , Williams Syndrome/psychology
15.
Brain ; 135(Pt 11): 3320-35, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23008234

ABSTRACT

The functional significance of adult neural stem and progenitor cells in hippocampal-dependent learning and memory has been well documented. Although adult neural stem and progenitor cells in the subventricular zone are known to migrate to, maintain and reorganize the olfactory bulb, it is less clear whether they are functionally required for other processes. Using a conditional transgenic mouse model, selective ablation of adult neural stem and progenitor cells in the subventricular zone induced a dramatic increase in morbidity and mortality of central nervous system disorders characterized by excitotoxicity-induced cell death accompanied by reactive inflammation, such as 4-aminopyridine-induced epilepsy and ischaemic stroke. To test the role of subventricular zone adult neural stem and progenitor cells in protecting central nervous system tissue from glutamatergic excitotoxicity, neurophysiological recordings of spontaneous excitatory postsynaptic currents from single medium spiny striatal neurons were measured on acute brain slices. Indeed, lipopolysaccharide-stimulated, but not unstimulated, subventricular zone adult neural stem and progenitor cells reverted the increased frequency and duration of spontaneous excitatory postsynaptic currents by secreting the endocannabinod arachidonoyl ethanolamide, a molecule that regulates glutamatergic tone through type 1 cannabinoid receptor (CB(1)) binding. In vivo restoration of cannabinoid levels, either by administration of the type 1 cannabinoid receptor agonist HU210 or the inhibitor of the principal catabolic enzyme fatty acid amide hydrolase, URB597, completely reverted the increased morbidity and mortality of adult neural stem and progenitor cell-ablated mice suffering from epilepsy and ischaemic stroke. Our results provide the first evidence that adult neural stem and progenitor cells located within the subventricular zone exert an 'innate' homeostatic regulatory role by protecting striatal neurons from glutamate-mediated excitotoxicity.


Subject(s)
Corpus Striatum/physiology , Glutamic Acid/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Neuroprotective Agents/metabolism , Stem Cells/physiology , 4-Aminopyridine/antagonists & inhibitors , Amidohydrolases/antagonists & inhibitors , Animals , Arachidonic Acids/biosynthesis , Arachidonic Acids/metabolism , Benzamides/pharmacology , Carbamates/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Endocannabinoids/biosynthesis , Endocannabinoids/metabolism , Epilepsy/metabolism , Epilepsy/mortality , Epilepsy/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Ganciclovir , Glutamic Acid/pharmacology , Lateral Ventricles/physiopathology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/drug effects , Polyunsaturated Alkamides , Stem Cells/drug effects , Stroke/metabolism , Stroke/mortality , Stroke/physiopathology
16.
Nat Commun ; 13(1): 161, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013317

ABSTRACT

Dravet syndrome is a severe epileptic encephalopathy caused primarily by haploinsufficiency of the SCN1A gene. Repetitive seizures can lead to endurable and untreatable neurological deficits. Whether this severe pathology is reversible after symptom onset remains unknown. To address this question, we generated a Scn1a conditional knock-in mouse model (Scn1a Stop/+) in which Scn1a expression can be re-activated on-demand during the mouse lifetime. Scn1a gene disruption leads to the development of seizures, often associated with sudden unexpected death in epilepsy (SUDEP) and behavioral alterations including hyperactivity, social interaction deficits and cognitive impairment starting from the second/third week of age. However, we showed that Scn1a gene re-activation when symptoms were already manifested (P30) led to a complete rescue of both spontaneous and thermic inducible seizures, marked amelioration of behavioral abnormalities and normalization of hippocampal fast-spiking interneuron firing. We also identified dramatic gene expression alterations, including those associated with astrogliosis in Dravet syndrome mice, that, accordingly, were rescued by Scn1a gene expression normalization at P30. Interestingly, regaining of Nav1.1 physiological level rescued seizures also in adult Dravet syndrome mice (P90) after months of repetitive attacks. Overall, these findings represent a solid proof-of-concept highlighting that disease phenotype reversibility can be achieved when Scn1a gene activity is efficiently reconstituted in brain cells.


Subject(s)
Cognitive Dysfunction/genetics , Epilepsies, Myoclonic/genetics , Hippocampus/metabolism , Interneurons/metabolism , NAV1.1 Voltage-Gated Sodium Channel/genetics , Sudden Unexpected Death in Epilepsy/prevention & control , Action Potentials/physiology , Animals , Cerebellum/metabolism , Cerebellum/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/prevention & control , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/physiopathology , Epilepsies, Myoclonic/prevention & control , Gene Knock-In Techniques , Genetic Therapy/methods , Hippocampus/physiopathology , Humans , Interneurons/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAV1.1 Voltage-Gated Sodium Channel/deficiency , Sudden Unexpected Death in Epilepsy/pathology
17.
Nat Commun ; 13(1): 7579, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36482070

ABSTRACT

The adult brain retains over life endogenous neural stem/precursor cells (eNPCs) within the subventricular zone (SVZ). Whether or not these cells exert physiological functions is still unclear. In the present work, we provide evidence that SVZ-eNPCs tune structural, electrophysiological, and behavioural aspects of striatal function via secretion of insulin-like growth factor binding protein-like 1 (IGFBPL1). In mice, selective ablation of SVZ-eNPCs or selective abrogation of IGFBPL1 determined an impairment of striatal medium spiny neuron morphology, a higher failure rate in GABAergic transmission mediated by fast-spiking interneurons, and striatum-related behavioural dysfunctions. We also found IGFBPL1 expression in the human SVZ, foetal and induced-pluripotent stem cell-derived NPCs. Finally, we found a significant correlation between SVZ damage, reduction of striatum volume, and impairment of information processing speed in neurological patients. Our results highlight the physiological role of adult SVZ-eNPCs in supporting cognitive functions by regulating striatal neuronal activity.


Subject(s)
Insulin-Like Growth Factor Binding Proteins , Lateral Ventricles , Neural Stem Cells , Tumor Suppressor Proteins , Animals , Humans , Mice , Cardiac Electrophysiology , Insulin-Like Growth Factor Binding Proteins/physiology , Neural Stem Cells/physiology , Tumor Suppressor Proteins/physiology , Lateral Ventricles/physiology
18.
Sci Immunol ; 7(67): eabl9929, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-34812647

ABSTRACT

The development of a tractable small animal model faithfully reproducing human coronavirus disease 2019 pathogenesis would arguably meet a pressing need in biomedical research. Thus far, most investigators have used transgenic mice expressing the human ACE2 in epithelial cells (K18-hACE2 transgenic mice) that are intranasally instilled with a liquid severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) suspension under deep anesthesia. Unfortunately, this experimental approach results in disproportionate high central nervous system infection leading to fatal encephalitis, which is rarely observed in humans and severely limits this model's usefulness. Here, we describe the use of an inhalation tower system that allows exposure of unanesthetized mice to aerosolized virus under controlled conditions. Aerosol exposure of K18-hACE2 transgenic mice to SARS-CoV-2 resulted in robust viral replication in the respiratory tract, anosmia, and airway obstruction but did not lead to fatal viral neuroinvasion. When compared with intranasal inoculation, aerosol infection resulted in a more pronounced lung pathology including increased immune infiltration, fibrin deposition, and a transcriptional signature comparable to that observed in SARS-CoV-2­infected patients. This model may prove useful for studies of viral transmission, disease pathogenesis (including long-term consequences of SARS-CoV-2 infection), and therapeutic interventions.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/physiopathology , Disease Models, Animal , Encephalitis, Viral/prevention & control , Keratin-18/genetics , Nasal Sprays , SARS-CoV-2/physiology , Administration, Inhalation , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19/virology , Encephalitis, Viral/mortality , Epithelial Cells/metabolism , Female , Humans , Keratin-18/metabolism , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Transcriptome , Virus Replication
19.
Hum Mol Genet ; 18(1): 105-17, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18829665

ABSTRACT

The GDI1 gene, responsible in human for X-linked non-specific mental retardation, encodes alphaGDI, a regulatory protein common to all GTPases of the Rab family. Its alteration, leading to membrane accumulation of different Rab GTPases, may affect multiple steps in neuronal intracellular traffic. Using electron microscopy and electrophysiology, we now report that lack of alphaGDI impairs several steps in synaptic vesicle (SV) biogenesis and recycling in the hippocampus. Alteration of the SV reserve pool (RP) and a 50% reduction in the total number of SV in adult synapses may be dependent on a defective endosomal-dependent recycling and may lead to the observed alterations in short-term plasticity. As predicted by the synaptic characteristics of the mutant mice, the short-term memory deficit, observed when using fear-conditioning protocols with short intervals between trials, disappeared when the Gdi1 mutants were allowed to have longer intervals between sessions. Likewise, previously observed deficits in radial maze learning could be corrected by providing less challenging pre-training. This implies that an intact RP of SVs is necessary for memory processing under challenging conditions in mice. The possibility to correct the learning deficit in mice may have clinical implication for future studies in human.


Subject(s)
Cognition , Guanine Nucleotide Dissociation Inhibitors/genetics , Learning , Mental Retardation, X-Linked/metabolism , Neuronal Plasticity , Synaptic Vesicles/physiology , Animals , Female , Guanine Nucleotide Dissociation Inhibitors/metabolism , Hippocampus/metabolism , Male , Memory , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Mental Retardation, X-Linked/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Synapses/genetics , Synapses/metabolism , Synaptic Vesicles/genetics
20.
Metabolism ; 116: 154463, 2021 03.
Article in English | MEDLINE | ID: mdl-33309713

ABSTRACT

OBJECTIVES: GDI1 gene encodes for αGDI, a protein controlling the cycling of small GTPases, reputed to orchestrate vesicle trafficking. Mutations in human GDI1 are responsible for intellectual disability (ID). In mice with ablated Gdi1, a model of ID, impaired working and associative short-term memory was recorded. This cognitive phenotype worsens if the deletion of αGDI expression is restricted to neurons. However, whether astrocytes, key homeostasis providing neuroglial cells, supporting neurons via aerobic glycolysis, contribute to this cognitive impairment is unclear. METHODS: We carried out proteomic analysis and monitored [18F]-fluoro-2-deoxy-d-glucose uptake into brain slices of Gdi1 knockout and wild type control mice. d-Glucose utilization at single astrocyte level was measured by the Förster Resonance Energy Transfer (FRET)-based measurements of cytosolic cyclic AMP, d-glucose and L-lactate, evoked by agonists selective for noradrenaline and L-lactate receptors. To test the role of astrocyte-resident processes in disease phenotype, we generated an inducible Gdi1 knockout mouse carrying the Gdi1 deletion only in adult astrocytes and conducted behavioural tests. RESULTS: Proteomic analysis revealed significant changes in astrocyte-resident glycolytic enzymes. Imaging [18F]-fluoro-2-deoxy-d-glucose revealed an increased d-glucose uptake in Gdi1 knockout tissue versus wild type control mice, consistent with the facilitated d-glucose uptake determined by FRET measurements. In mice with Gdi1 deletion restricted to astrocytes, a selective and significant impairment in working memory was recorded, which was rescued by inhibiting glycolysis by 2-deoxy-d-glucose injection. CONCLUSIONS: These results reveal a new astrocyte-based mechanism in neurodevelopmental disorders and open a novel therapeutic opportunity of targeting aerobic glycolysis, advocating a change in clinical practice.


Subject(s)
Deoxyglucose/pharmacology , Glycolysis/drug effects , Guanine Nucleotide Dissociation Inhibitors/genetics , Intellectual Disability/genetics , Memory Disorders/prevention & control , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Deoxyglucose/therapeutic use , Down-Regulation/drug effects , Glucose/metabolism , Guanine Nucleotide Dissociation Inhibitors/deficiency , Intellectual Disability/drug therapy , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Maze Learning/drug effects , Memory/drug effects , Memory Disorders/genetics , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL