Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 7(1): 140, 2024 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-38291108

RESUMO

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.


Assuntos
Ceruloplasmina , Distúrbios do Metabolismo do Ferro , Doenças Neurodegenerativas , Proteoma , Adulto , Humanos , Animais , Camundongos , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Proteoma/metabolismo , Doenças Raras , Resíduos Industriais
2.
Nat Commun ; 13(1): 7579, 2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36482070

RESUMO

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.


Assuntos
Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Ventrículos Laterais , Células-Tronco Neurais , Proteínas Supressoras de Tumor , Animais , Humanos , Camundongos , Eletrofisiologia Cardíaca , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Células-Tronco Neurais/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Ventrículos Laterais/fisiologia
3.
Nat Commun ; 13(1): 161, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013317

RESUMO

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.


Assuntos
Disfunção Cognitiva/genética , Epilepsias Mioclônicas/genética , Hipocampo/metabolismo , Interneurônios/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Morte Súbita Inesperada na Epilepsia/prevenção & controle , Potenciais de Ação/fisiologia , Animais , Cerebelo/metabolismo , Cerebelo/fisiopatologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/prevenção & controle , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/fisiopatologia , Epilepsias Mioclônicas/prevenção & controle , Técnicas de Introdução de Genes , Terapia Genética/métodos , Hipocampo/fisiopatologia , Humanos , Interneurônios/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1/deficiência , Morte Súbita Inesperada na Epilepsia/patologia
4.
Hum Mol Genet ; 31(9): 1389-1406, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-34761259

RESUMO

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.


Assuntos
Transtorno do Espectro Autista , Deficiência Intelectual , Animais , Transtorno do Espectro Autista/genética , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Deficiência Intelectual/genética , Camundongos , Mutação , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
5.
Sci Immunol ; 7(67): eabl9929, 2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-34812647

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2/genética , COVID-19/fisiopatologia , Modelos Animais de Doenças , Encefalite Viral/prevenção & controle , Queratina-18/genética , Sprays Nasais , SARS-CoV-2/fisiologia , Administração por Inalação , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , COVID-19/imunologia , COVID-19/virologia , Encefalite Viral/mortalidade , Células Epiteliais/metabolismo , Feminino , Humanos , Queratina-18/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Transcriptoma , Replicação Viral
6.
Mol Psychiatry ; 26(11): 6531-6549, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34035473

RESUMO

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.


Assuntos
Espinhas Dendríticas , Deficiência Intelectual , Proteínas rab de Ligação ao GTP , Animais , Cálcio , Espinhas Dendríticas/fisiologia , Camundongos , Plasticidade Neuronal , Neurônios/fisiologia , Receptores de Glutamato/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia
7.
Metabolism ; 116: 154463, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33309713

RESUMO

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.


Assuntos
Desoxiglucose/farmacologia , Glicólise/efeitos dos fármacos , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Deficiência Intelectual/genética , Transtornos da Memória/prevenção & controle , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Desoxiglucose/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Glucose/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/deficiência , Deficiência Intelectual/tratamento farmacológico , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Transtornos da Memória/genética , Camundongos , Camundongos Knockout
8.
Mol Neurobiol ; 56(7): 4838-4854, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30402709

RESUMO

MeCP2 is a fundamental protein associated with several neurological disorders, including Rett syndrome. It is considered a multifunctional factor with a prominent role in regulating chromatin structure; however, a full comprehension of the consequences of its deficiency is still lacking. Here, we characterize a novel mouse model of Mecp2 bearing the human mutation Y120D, which is localized in the methyl-binding domain. As most models of Mecp2, the Mecp2Y120D mouse develops a severe Rett-like phenotype. This mutation alters the interaction of the protein with chromatin, but surprisingly, it also impairs its association with corepressors independently on the involved interacting domains. These features, which become overt mainly in the mature brain, cause a more accessible and transcriptionally active chromatin structure; conversely, in the Mecp2-null brain, we find a less accessible and transcriptionally inactive chromatin. By demonstrating that different MECP2 mutations can produce concordant neurological phenotypes but discordant molecular features, we highlight the importance of considering personalized approaches for the treatment of Rett syndrome.


Assuntos
Comportamento Animal , Técnicas de Introdução de Genes , Proteína 2 de Ligação a Metil-CpG/metabolismo , Medicina de Precisão , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cromatina/metabolismo , Feminino , Humanos , Longevidade , Masculino , Memória de Curto Prazo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutação/genética , Neurônios/metabolismo , Fenótipo , Síndrome de Rett
9.
Hum Mol Genet ; 28(1): 124-132, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30239779

RESUMO

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.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteína P0 da Mielina/genética , Proteína P0 da Mielina/fisiologia , Animais , Axônios/metabolismo , Membrana Celular/fisiologia , Códon sem Sentido , Doenças Desmielinizantes/genética , Feminino , Técnicas de Introdução de Genes , Humanos , Masculino , Camundongos Endogâmicos BALB C , Mutação , Fenótipo , Transporte Proteico/genética , Células de Schwann/metabolismo
10.
J Neuroinflammation ; 15(1): 58, 2018 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-29475438

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Locomoção/fisiologia , Células-Tronco Neurais/patologia , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/patologia , Medula Espinal/patologia , Animais , Proliferação de Células/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia
11.
EMBO Mol Med ; 10(1): 91-106, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29183916

RESUMO

Aceruloplasminemia is a monogenic disease caused by mutations in the ceruloplasmin gene that result in loss of protein ferroxidase activity. Ceruloplasmin plays a role in iron homeostasis, and its activity impairment leads to iron accumulation in liver, pancreas, and brain. Iron deposition promotes diabetes, retinal degeneration, and progressive neurodegeneration. Current therapies mainly based on iron chelation, partially control systemic iron deposition but are ineffective on neurodegeneration. We investigated the potential of ceruloplasmin replacement therapy in reducing the neurological pathology in the ceruloplasmin-knockout (CpKO) mouse model of aceruloplasminemia. CpKO mice were intraperitoneal administered for 2 months with human ceruloplasmin that was able to enter the brain inducing replacement of the protein levels and rescue of ferroxidase activity. Ceruloplasmin-treated mice showed amelioration of motor incoordination that was associated with diminished loss of Purkinje neurons and reduced brain iron deposition, in particular in the choroid plexus. Computational analysis showed that ceruloplasmin-treated CpKO mice share a similar pattern with wild-type animals, highlighting the efficacy of the therapy. These data suggest that enzyme replacement therapy may be a promising strategy for the treatment of aceruloplasminemia.


Assuntos
Ceruloplasmina/deficiência , Ceruloplasmina/uso terapêutico , Distúrbios do Metabolismo do Ferro/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Ceruloplasmina/administração & dosagem , Ceruloplasmina/metabolismo , Ceruloplasmina/farmacocinética , Terapia Enzimática , Feminino , Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Masculino , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia
12.
Small GTPases ; 9(1-2): 145-157, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-28146371

RESUMO

Neurons are highly polarized cells that exhibit one of the more complex morphology and function. Neuronal intracellular trafficking plays a key role in dictating the directionality and specificity of vesicle formation, transport and fusion, allowing the transmission of information in sophisticate cellular network. Thus, the integrity of protein trafficking and spatial organization is especially important in neuronal cells. RAB proteins, small monomeric GTPases belonging to the RAS superfamily, spatially and temporally orchestrate specific vesicular trafficking steps. In this review we summarise the known roles of RAB GTPases involved in the maintenance of neuronal vesicular trafficking in the central nervous system. In particular, we discriminate the axonal pre-synaptic trafficking and dendritic post-synaptic trafficking, to better underlie how a correct orchestration of vesicle movement is necessary to maintain neuronal polarity and then, to permit an accurate architecture and functionality of synaptic activity.


Assuntos
Sinapses/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Transporte Biológico , Dendritos/metabolismo , Humanos
13.
Cereb Cortex ; 27(11): 5369-5384, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28968657

RESUMO

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.


Assuntos
Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Deficiência Intelectual/tratamento farmacológico , Deficiência Intelectual/metabolismo , Proteínas de Membrana/deficiência , Proteínas do Tecido Nervoso/deficiência , Psicotrópicos/farmacologia , Receptores de AMPA/metabolismo , Regulação Alostérica , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Deficiência Intelectual/patologia , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , Ligação Proteica/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/ultraestrutura , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
14.
Hum Mol Genet ; 26(23): 4699-4714, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973667

RESUMO

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.


Assuntos
Retardo Mental Ligado ao Cromossomo X/genética , Mutação de Sentido Incorreto , Sinapsinas/genética , Vesículas Sinápticas/genética , Animais , Sequência de Bases , Humanos , Retardo Mental Ligado ao Cromossomo X/metabolismo , Camundongos , Camundongos Knockout , Mutação , Neurogênese/genética , Plasticidade Neuronal/genética , Neurônios/metabolismo , Linhagem , Terminações Pré-Sinápticas/metabolismo , Cultura Primária de Células , Transporte Proteico , Sinapsinas/metabolismo , Transmissão Sináptica/genética , Vesículas Sinápticas/metabolismo
15.
Parkinsonism Relat Disord ; 44: 142-146, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28851564

RESUMO

BACKGROUND: RAB39B pathogenic variants cause X-linked Parkinsonism associated with Intellectual Disability, known as Waisman syndrome, a very rare disorder that has been mainly identified through exome sequencing in large Parkinson's disease cohorts. In this study we searched for pathogenic variants in RAB39B in two Italian families affected by X-linked early-onset Parkinsonism and Intellectual Disability. METHODS: Three patients received neurological evaluation and underwent RAB39B sequencing. RESULTS: Two novel RAB39B frameshift variants were found to result in the absence of RAB39B protein (family 1: c.137dupT; family 2: c.371delA). Patients showed unilateral rest tremor and bradykinesia; one of them also displayed an early-onset postural tremor. Paramagnetic substance deposition in the substantia nigra, globus pallidi, red nucleus, putamen and pulvinar was assessed by brain imaging. Two patients also showed moderate calcification of globus pallidi. CONCLUSION: In this study we highlight the evidence that X-linked early-onset Parkinsonism associated with Intellectual Disability occurs as a pattern of clinical and neuroimaging features attributable to RAB39B pathogenic variants.


Assuntos
Doenças dos Gânglios da Base/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Deficiência Intelectual/genética , Doença de Parkinson/genética , Proteínas rab de Ligação ao GTP/genética , Idoso , Humanos , Masculino , Pessoa de Meia-Idade , Mosaicismo , Mutação , Linhagem
16.
Neuroscience ; 344: 346-359, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28057534

RESUMO

RAB-GDP dissociation inhibitor 1 (GDI1) loss-of-function mutations are responsible for a form of non-specific X-linked Intellectual Disability (XLID) where the only clinical feature is cognitive impairment. GDI1 patients are impaired in specific aspects of executive functions and conditioned response, which are controlled by fronto-striatal circuitries. Previous molecular and behavioral characterization of the Gdi1-null mouse revealed alterations in the total number/distribution of hippocampal and cortical synaptic vesicles as well as hippocampal short-term synaptic plasticity, and memory deficits. In this study, we employed cognitive protocols with high translational validity to human condition that target the functionality of cortico-striatal circuitry such as attention and stimulus selection ability with progressive degree of complexity. We previously showed that Gdi1-null mice are impaired in some hippocampus-dependent forms of associative learning assessed by aversive procedures. Here, using appetitive-conditioning procedures we further investigated associative learning deficits sustained by the fronto-striatal system. We report that Gdi1-null mice are impaired in attention and associative learning processes, which are a key part of the cognitive impairment observed in XLID patients.


Assuntos
Lobo Frontal/fisiopatologia , Inibidores de Dissociação do Nucleotídeo Guanina/deficiência , Deficiência Intelectual/fisiopatologia , Neostriado/fisiopatologia , Tonsila do Cerebelo/diagnóstico por imagem , Tonsila do Cerebelo/fisiopatologia , Animais , Aprendizagem por Associação/fisiologia , Atenção/fisiologia , Condicionamento Psicológico/fisiologia , Discriminação Psicológica/fisiologia , Modelos Animais de Doenças , Dopamina/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Lobo Frontal/diagnóstico por imagem , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Inibição Psicológica , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/psicologia , Masculino , Camundongos Knockout , Neostriado/diagnóstico por imagem , Vias Neurais/diagnóstico por imagem , Vias Neurais/fisiopatologia , Distribuição Aleatória , Vesículas Sinápticas/metabolismo , Percepção do Tempo/fisiologia , Técnicas de Cultura de Tecidos
17.
Sci Rep ; 7: 40136, 2017 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28074903

RESUMO

Adenosine Deaminase (ADA) deficiency is an autosomal recessive variant of severe combined immunodeficiency (SCID) caused by systemic accumulation of ADA substrates. Neurological and behavioral abnormalities observed in ADA-SCID patients surviving after stem cell transplantation or gene therapy represent an unresolved enigma in the field. We found significant neurological and cognitive alterations in untreated ADA-SCID patients as well as in two groups of patients after short- and long-term enzyme replacement therapy with PEG-ADA. These included motor dysfunction, EEG alterations, sensorineural hypoacusia, white matter and ventricular alterations in MRI as well as a low mental development index or IQ. Ada-deficient mice were significantly less active and showed anxiety-like behavior. Molecular and metabolic analyses showed that this phenotype coincides with metabolic alterations and aberrant adenosine receptor signaling. PEG-ADA treatment corrected metabolic adenosine-based alterations, but not cellular and signaling defects, indicating an intrinsic nature of the neurological and behavioral phenotype in ADA deficiency.


Assuntos
Adenosina Desaminase/deficiência , Adenosina/metabolismo , Encéfalo/metabolismo , Doenças do Sistema Nervoso/fisiopatologia , Animais , Comportamento , Comportamento Animal , Humanos , Camundongos , Doenças do Sistema Nervoso/patologia
18.
Mol Neurobiol ; 54(4): 2458-2468, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26971292

RESUMO

X-linked non-syndromic intellectual disability (XLID) is a common mental disorder recognized by cognitive and behavioral deficits. Mutations in the brain-specific αGDI, shown to alter a subset of RAB GTPases redistribution in cells, are linked to XLID, likely via changes in vesicle traffic in neurons. Here, we show directly that isolated XLID mice astrocytes, devoid of pathologic tissue environment, exhibit vesicle mobility deficits. Contrary to previous studies, we show that astrocytes express two GDI proteins. The siRNA-mediated suppression of expression of αGDI especially affected vesicle dynamics. A similar defect was recorded in astrocytes from the Gdi1 -/Y mouse model of XLID and in astrocytes with recombinant mutated human XLID αGDI. Endolysosomal vesicles studied here are involved in the release of gliosignaling molecules as well as in regulating membrane receptor density; thus, the observed changes in astrocytic vesicle mobility may, over the long time-course, profoundly affect signaling capacity of these cells, which optimize neural activity.


Assuntos
Astrócitos/metabolismo , Vesículas Citoplasmáticas/metabolismo , Genes Ligados ao Cromossomo X , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Deficiência Intelectual/genética , Animais , Astrócitos/patologia , Modelos Animais de Doenças , Endossomos/metabolismo , Inativação Gênica , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Lisossomos/metabolismo , Masculino , Camundongos , Mutação/genética , Ratos , Transfecção
19.
EMBO Mol Med ; 8(12): 1438-1454, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27799291

RESUMO

Charcot-Marie-Tooth (CMT) neuropathies are highly heterogeneous disorders caused by mutations in more than 70 genes, with no available treatment. Thus, it is difficult to envisage a single suitable treatment for all pathogenetic mechanisms. Axonal Neuregulin 1 (Nrg1) type III drives Schwann cell myelination and determines myelin thickness by ErbB2/B3-PI3K-Akt signaling pathway activation. Nrg1 type III is inhibited by the α-secretase Tace, which negatively regulates PNS myelination. We hypothesized that modulation of Nrg1 levels and/or secretase activity may constitute a unifying treatment strategy for CMT neuropathies with focal hypermyelination as it could restore normal levels of myelination. Here we show that in vivo delivery of Niaspan, a FDA-approved drug known to enhance TACE activity, efficiently rescues myelination in the Mtmr2-/- mouse, a model of CMT4B1 with myelin outfoldings, and in the Pmp22+/- mouse, which reproduces HNPP (hereditary neuropathy with liability to pressure palsies) with tomacula. Importantly, we also found that Niaspan reduces hypermyelination of Vim (vimentin)-/- mice, characterized by increased Nrg1 type III and Akt activation, thus corroborating the hypothesis that Niaspan treatment downregulates Nrg1 type III signaling.


Assuntos
Proteína ADAM17/metabolismo , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Doença de Charcot-Marie-Tooth/patologia , Fármacos Neuroprotetores/administração & dosagem , Niacina/administração & dosagem , Complexo Vitamínico B/administração & dosagem , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Resultado do Tratamento
20.
Sci Rep ; 6: 34877, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27713499

RESUMO

During brain development, the small GTPases Rac1/Rac3 play key roles in neuronal migration, neuritogenesis, synaptic formation and plasticity, via control of actin cytoskeleton dynamic. Their activity is positively and negatively regulated by GEFs and GAPs molecules, respectively. However their in vivo roles are poorly known. The ArhGAP15 gene, coding for a Rac-specific GAP protein, is expressed in both excitatory and inhibitory neurons of the adult hippocampus, and its loss results in the hyperactivation of Rac1/Rac3. In the CA3 and dentate gyrus (DG) regions of the ArhGAP15 mutant hippocampus the CR+, PV+ and SST+ inhibitory neurons are reduced in number, due to reduced efficiency and directionality of their migration, while pyramidal neurons are unaffected. Loss of ArhGAP15 alters neuritogenesis and the balance between excitatory and inhibitory synapses, with a net functional result consisting in increased spike frequency and bursts, accompanied by poor synchronization. Thus, the loss of ArhGAP15 mainly impacts on interneuron-dependent inhibition. Adult ArhGAP15-/- mice showed defective hippocampus-dependent functions such as working and associative memories. These findings indicate that a normal architecture and function of hippocampal inhibitory neurons is essential for higher hippocampal functions, and is exquisitely sensitive to ArhGAP15-dependent modulation of Rac1/Rac3.


Assuntos
Transtornos Cognitivos/genética , Proteínas Ativadoras de GTPase/metabolismo , Hipocampo/fisiopatologia , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Comportamento Animal/fisiologia , Movimento Celular/genética , Células Cultivadas , Transtornos Cognitivos/etiologia , Feminino , Proteínas Ativadoras de GTPase/genética , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/patologia , Interneurônios/patologia , Masculino , Memória de Curto Prazo/fisiologia , Camundongos Mutantes , Neurônios/patologia , Neuropeptídeos/genética , Ratos , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...