Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Front Mol Neurosci ; 17: 1399953, 2024.
Article En | MEDLINE | ID: mdl-38756706

Over the years, advancements in modeling neurological diseases have revealed innovative strategies aimed at gaining deeper insights and developing more effective treatments for these complex conditions. However, these progresses have recently been overshadowed by an increasing number of failures in clinical trials, raising doubts about the reliability and translatability of this type of disease modeling. This mini-review does not aim to provide a comprehensive overview of the current state-of-the-art in disease mouse modeling. Instead, it offers a brief excursus over some recent approaches in modeling neurological diseases to pinpoint a few intriguing strategies applied in the field that may serve as sources of inspiration for improving currently available animal models. In particular, we aim to guide the reader toward the potential success of adopting a more orthogonal approach in the study of human diseases.

2.
Blood ; 143(14): 1399-1413, 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38194688

ABSTRACT: SETBP1 mutations are found in various clonal myeloid disorders. However, it is unclear whether they can initiate leukemia, because SETBP1 mutations typically appear as later events during oncogenesis. To answer this question, we generated a mouse model expressing mutated SETBP1 in hematopoietic tissue: this model showed profound alterations in the differentiation program of hematopoietic progenitors and developed a myeloid neoplasm with megakaryocytic dysplasia, splenomegaly, and bone marrow fibrosis, prompting us to investigate SETBP1 mutations in a cohort of 36 triple-negative primary myelofibrosis (TN-PMF) cases. We identified 2 distinct subgroups, one carrying SETBP1 mutations and the other completely devoid of somatic variants. Clinically, a striking difference in disease aggressiveness was noted, with patients with SETBP1 mutation showing a much worse clinical course. In contrast to myelodysplastic/myeloproliferative neoplasms, in which SETBP1 mutations are mostly found as a late clonal event, single-cell clonal hierarchy reconstruction in 3 patients with TN-PMF from our cohort revealed SETBP1 to be a very early event, suggesting that the phenotype of the different SETBP1+ disorders may be shaped by the opposite hierarchy of the same clonal SETBP1 variants.


Hematopoietic System , Myelodysplastic-Myeloproliferative Diseases , Myeloproliferative Disorders , Primary Myelofibrosis , Animals , Mice , Humans , Primary Myelofibrosis/genetics , Myeloproliferative Disorders/genetics , Mutation , Carrier Proteins/genetics , Nuclear Proteins/genetics
3.
Nat Commun ; 14(1): 3212, 2023 06 03.
Article En | MEDLINE | ID: mdl-37270547

Within the chromatin, distal elements interact with promoters to regulate specific transcriptional programs. Histone acetylation, interfering with the net charges of the nucleosomes, is a key player in this regulation. Here, we report that the oncoprotein SET is a critical determinant for the levels of histone acetylation within enhancers. We disclose that a condition in which SET is accumulated, the severe Schinzel-Giedion Syndrome (SGS), is characterized by a failure in the usage of the distal regulatory regions typically employed during fate commitment. This is accompanied by the usage of alternative enhancers leading to a massive rewiring of the distal control of the gene transcription. This represents a (mal)adaptive mechanism that, on one side, allows to achieve a certain degree of differentiation, while on the other affects the fine and corrected maturation of the cells. Thus, we propose the differential in cis-regulation as a contributing factor to the pathological basis of SGS and possibly other the SET-related disorders in humans.


Enhancer Elements, Genetic , Histones , Humans , Histones/genetics , Histones/metabolism , Enhancer Elements, Genetic/genetics , Cell Differentiation/genetics , Chromatin/genetics , Promoter Regions, Genetic/genetics
4.
Mol Autism ; 14(1): 20, 2023 06 01.
Article En | MEDLINE | ID: mdl-37264456

BACKGROUND: Neurodevelopmental disorders (NDDs) are heterogeneous conditions due to alterations of a variety of molecular mechanisms and cell dysfunctions. SETD5 haploinsufficiency leads to NDDs due to chromatin defects. Epigenetic basis of NDDs has been reported in an increasing number of cases while mitochondrial dysfunctions are more common within NDD patients than in the general population. METHODS: We investigated in vitro neural stem cells as well as the brain of the Setd5 haploinsufficiency mouse model interrogating its transcriptome, analyzing mitochondrial structure, biochemical composition, and dynamics, as well as mitochondrial functionality. RESULTS: Mitochondrial impairment is facilitated by transcriptional aberrations originated by the decrease of the SETD5 enzyme. Low levels of SETD5 resulted in fragmented mitochondria, reduced mitochondrial membrane potential, and ATP production both in neural precursors and neurons. Mitochondria were also mislocalized in mutant neurons, with reduced organelles within neurites and synapses. LIMITATIONS: We found several defects in the mitochondrial compartment; however, we can only speculate about their position in the hierarchy of the pathological mechanisms at the basis of the disease. CONCLUSIONS: Our study explores the interplay between chromatin regulation and mitochondria functions as a possible important aspect of SETD5-associated NDD pathophysiology. Our data, if confirmed in patient context, suggest that the mitochondrial activity and dynamics may represent new therapeutic targets for disorders associated with the loss of SETD5.


Haploinsufficiency , Neural Stem Cells , Mice , Animals , Humans , Neurons/metabolism , Mitochondria/metabolism , Neural Stem Cells/metabolism , Chromatin/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism
6.
Sci Adv ; 8(31): eabn3986, 2022 08 05.
Article En | MEDLINE | ID: mdl-35921410

Current therapies remain unsatisfactory in preventing the recurrence of glioblastoma multiforme (GBM), which leads to poor patient survival. By rational engineering of the transcription factor SOX2, a key promoter of GBM malignancy, together with the Kruppel-associated box and DNA methyltransferase3A/L catalytic domains, we generated a synthetic repressor named SOX2 epigenetic silencer (SES), which induces the transcriptional silencing of its original targets. By doing so, SES kills both glioma cell lines and patient-derived cancer stem cells in vitro and in vivo. SES expression, through local viral delivery in mouse xenografts, induces strong regression of human tumors and survival rescue. Conversely, SES is not harmful to neurons and glia, also thanks to a minimal promoter that restricts its expression in mitotically active cells, rarely present in the brain parenchyma. Collectively, SES produces a significant silencing of a large fraction of the SOX2 transcriptional network, achieving high levels of efficacy in repressing aggressive brain tumors.


Brain Neoplasms , Glioblastoma , Glioma , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Epigenesis, Genetic , Glioblastoma/metabolism , Glioma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
7.
Sci Immunol ; 7(67): eabl9929, 2022 Jan 28.
Article En | MEDLINE | ID: mdl-34812647

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.


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
8.
Int J Mol Sci ; 24(1)2022 Dec 22.
Article En | MEDLINE | ID: mdl-36613611

Haploinsufficiency of the SETD5 gene, encoding a SET domain-containing histone methyltransferase, has been identified as a cause of intellectual disability and Autism Spectrum Disorder (ASD). Recently, the zebrafish has emerged as a valuable model to study neurodevelopmental disorders because of its genetic tractability, robust behavioral traits and amenability to high-throughput drug screening. To model human SETD5 haploinsufficiency, we generated zebrafish setd5 mutants using the CRISPR/Cas9 technology and characterized their morphological, behavioral and molecular phenotypes. According to our observation that setd5 is expressed in adult zebrafish brain, including those areas controlling social behavior, we found that setd5 heterozygous mutants exhibit defective aggregation and coordination abilities required for shoaling interactions, as well as indifference to social stimuli. Interestingly, impairment in social interest is rescued by risperidone, an antipsychotic drug used to treat behavioral traits in ASD individuals. The molecular analysis underscored the downregulation of genes encoding proteins involved in the synaptic structure and function in the adult brain, thus suggesting that brain hypo-connectivity could be responsible for the social impairments of setd5 mutant fishes. The zebrafish setd5 mutants display ASD-like features and are a promising setd5 haploinsufficiency model for drug screening aimed at reversing the behavioral phenotypes.


Autism Spectrum Disorder , Methyltransferases , Animals , Humans , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Brain/metabolism , CRISPR-Cas Systems , Methyltransferases/genetics , Methyltransferases/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Social Behavior
9.
Biomolecules ; 11(11)2021 11 17.
Article En | MEDLINE | ID: mdl-34827709

The advent of next-generation sequencing (NGS) is heavily changing both the diagnosis of human conditions and basic biological research. It is now possible to dig deep inside the genome of hundreds of thousands or even millions of people and find both common and rare genomic variants and to perform detailed phenotypic characterizations of both physiological organs and experimental models. Recent years have seen the introduction of multiple techniques using NGS to profile transcription, DNA and chromatin modifications, protein binding, etc., that are now allowing us to profile cells in bulk or even at a single-cell level. Although rare and ultra-rare diseases only affect a few people, each of these diseases represent scholarly cases from which a great deal can be learned about the pathological and physiological function of genes, pathways, and mechanisms. Therefore, for rare diseases, state-of-the-art investigations using NGS have double valence: their genomic cause (new variants) and the characterize the underlining the mechanisms associated with them (discovery of gene function) can be found. In a non-exhaustive manner, this review will outline the main usage of NGS-based techniques for the diagnosis and characterization of neurodevelopmental disorders (NDDs), under whose umbrella many rare and ultra-rare diseases fall.


High-Throughput Nucleotide Sequencing , Rare Diseases , Humans , Sequence Analysis, DNA
10.
Nat Commun ; 12(1): 4050, 2021 06 30.
Article En | MEDLINE | ID: mdl-34193871

The investigation of genetic forms of juvenile neurodegeneration could shed light on the causative mechanisms of neuronal loss. Schinzel-Giedion syndrome (SGS) is a fatal developmental syndrome caused by mutations in the SETBP1 gene, inducing the accumulation of its protein product. SGS features multi-organ involvement with severe intellectual and physical deficits due, at least in part, to early neurodegeneration. Here we introduce a human SGS model that displays disease-relevant phenotypes. We show that SGS neural progenitors exhibit aberrant proliferation, deregulation of oncogenes and suppressors, unresolved DNA damage, and resistance to apoptosis. Mechanistically, we demonstrate that high SETBP1 levels inhibit P53 function through the stabilization of SET, which in turn hinders P53 acetylation. We find that the inheritance of unresolved DNA damage in SGS neurons triggers the neurodegenerative process that can be alleviated either by PARP-1 inhibition or by NAD + supplementation. These results implicate that neuronal death in SGS originates from developmental alterations mainly in safeguarding cell identity and homeostasis.


Abnormalities, Multiple/pathology , Carrier Proteins/metabolism , Craniofacial Abnormalities/pathology , DNA Damage , Hand Deformities, Congenital/pathology , Heredodegenerative Disorders, Nervous System/pathology , Intellectual Disability/pathology , Mutation , Nails, Malformed/pathology , Neural Stem Cells/pathology , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Carrier Proteins/genetics , Cells, Cultured , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Hand Deformities, Congenital/genetics , Hand Deformities, Congenital/metabolism , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Nails, Malformed/genetics , Nails, Malformed/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/genetics , Organoids
11.
Front Cell Dev Biol ; 9: 641410, 2021.
Article En | MEDLINE | ID: mdl-33708771

TBL1XR1 gene is associated with multiple developmental disorders presenting several neurological aspects. The relative protein is involved in the modulation of important cellular pathways and master regulators of transcriptional output, including nuclear receptor repressors, Wnt signaling, and MECP2 protein. However, TBL1XR1 mutations (including complete loss of its functions) have not been experimentally studied in a neurological context, leaving a knowledge gap in the mechanisms at the basis of the diseases. Here, we show that Tbl1xr1 knock-out mice exhibit behavioral and neuronal abnormalities. Either the absence of TBL1XR1 or its point mutations interfering with stability/regulation of NCOR complex induced decreased proliferation and increased differentiation in neural progenitors. We suggest that this developmental unbalance is due to a failure in the regulation of the MAPK cascade. Taken together, our results broaden the molecular and functional aftermath of TBL1XR1 deficiency associated with human disorders.

12.
J Neurosci ; 40(37): 7013-7026, 2020 09 09.
Article En | MEDLINE | ID: mdl-32801157

Sulfotransferase 4A1 (SULT4A1) is a cytosolic sulfotransferase that is highly conserved across species and extensively expressed in the brain. However, the biological function of SULT4A1 is unclear. SULT4A1 has been implicated in several neuropsychiatric disorders, such as Phelan-McDermid syndrome and schizophrenia. Here, we investigate the role of SULT4A1 within neuron development and function. Our data demonstrate that SULT4A1 modulates neuronal branching complexity and dendritic spines formation. Moreover, we show that SULT4A1, by negatively regulating the catalytic activity of Pin1 toward PSD-95, facilitates NMDAR synaptic expression and function. Finally, we demonstrate that the pharmacological inhibition of Pin1 reverses the pathologic phenotypes of neurons knocked down by SULT4A1 by specifically restoring dendritic spine density and rescuing NMDAR-mediated synaptic transmission. Together, these findings identify SULT4A1 as a novel player in neuron development and function by modulating dendritic morphology and synaptic activity.SIGNIFICANCE STATEMENT Sulfotransferase 4A1 (SULT4A1) is a brain-specific sulfotransferase highly expressed in neurons. Different evidence has suggested that SULT4A1 has an important role in neuronal function and that SULT4A1 altered expression might represent a contributing factor in multiple neurodevelopmental disorders. However, the function of SULT4A1 in the mammalian brain is still unclear. Here, we demonstrate that SULT4A1 is highly expressed at postsynaptic sites where it sequesters Pin1, preventing its negative action on synaptic transmission. This study reveals a novel role of SULT4A1 in the modulation of NMDA receptor activity and strongly contributes to explaining the neuronal dysfunction observed in patients carrying deletions of SULTA41 gene.


Disks Large Homolog 4 Protein/metabolism , Neurogenesis , Receptors, N-Methyl-D-Aspartate/metabolism , Sulfotransferases/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Dendritic Spines/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Rats , Sulfotransferases/genetics , Synapses/physiology , Synaptic Transmission
13.
Cell Rep ; 29(5): 1130-1146.e8, 2019 10 29.
Article En | MEDLINE | ID: mdl-31665629

Tetraspanins are a class of evolutionarily conserved transmembrane proteins with 33 members identified in mammals that have the ability to organize specific membrane domains, named tetraspanin-enriched microdomains (TEMs). Despite the relative abundance of different tetraspanins in the CNS, few studies have explored their role at synapses. Here, we investigate the function of TSPAN5, a member of the tetraspanin superfamily for which mRNA transcripts are found at high levels in the mouse brain. We demonstrate that TSPAN5 is localized in dendritic spines of pyramidal excitatory neurons and that TSPAN5 knockdown induces a dramatic decrease in spine number because of defects in the spine maturation process. Moreover, we show that TSPAN5 interacts with the postsynaptic adhesion molecule neuroligin-1, promoting its correct surface clustering. We propose that membrane compartmentalization by tetraspanins represents an additional mechanism for regulating excitatory synapses.


Cell Adhesion Molecules, Neuronal/metabolism , Dendritic Spines/metabolism , Membrane Microdomains/metabolism , Tetraspanins/chemistry , Tetraspanins/metabolism , Animals , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Protein Binding , Pyramidal Cells/metabolism , Rats, Wistar , Synapses/metabolism
14.
Neuron ; 104(2): 271-289.e13, 2019 10 23.
Article En | MEDLINE | ID: mdl-31515109

Mutations in one SETD5 allele are genetic causes of intellectual disability and autistic spectrum disorders. However, the mechanisms by which SETD5 regulates brain development and function remain largely elusive. Herein, we found that Setd5 haploinsufficiency impairs the proliferative dynamics of neural progenitors and synaptic wiring of neurons, ultimately resulting in behavioral deficits in mice. Mechanistically, Setd5 inactivation in neural stem cells, zebrafish, and mice equally affects genome-wide levels of H3K36me3 on active gene bodies. Notably, we demonstrated that SETD5 directly deposits H3K36me3, which is essential to allow on-time RNA elongation dynamics. Hence, Setd5 gene loss leads to abnormal transcription, with impaired RNA maturation causing detrimental effects on gene integrity and splicing. These findings identify SETD5 as a fundamental epigenetic enzyme controlling the transcriptional landscape in neural progenitors and their derivatives and illuminate the molecular events that connect epigenetic defects with neuronal dysfunctions at the basis of related human diseases.


Brain/embryology , Chromatin/metabolism , Gene Expression Regulation, Developmental/genetics , Histone Code/genetics , Methyltransferases/genetics , Zebrafish Proteins/physiology , Animals , Behavior, Animal , Brain/metabolism , Chromatin Immunoprecipitation Sequencing , Cognition , Epigenesis, Genetic , Histone Methyltransferases/genetics , Methyltransferases/physiology , Mice , Mutation , Neural Stem Cells/metabolism , RNA Splicing/genetics , RNA-Seq , Social Behavior , Transcription Elongation, Genetic , Zebrafish , Zebrafish Proteins/genetics
15.
Front Genet ; 10: 1291, 2019.
Article En | MEDLINE | ID: mdl-31998360

Post-translational methylation of H3 lysine 36 (H3K36) is an important epigenetic marker that majorly contributes to the functionality of the chromatin. This mark is interpreted by the cell in several crucial biological processes including gene transcription and DNA methylation. The homeostasis of H3K36 methylation is finely regulated by different enzyme classes which, when impaired, lead to a plethora of diseases; ranging from multi-organ syndromes to cancer, to pure neurological diseases often associated with brain development. This mini-review summarizes current knowledge on these important epigenetic signals with emphasis on the molecular mechanisms that (i) regulate their abundance, (ii) are influenced by H3K36 methylation, and (iii) the associated diseases.

16.
Nat Commun ; 9(1): 2192, 2018 06 06.
Article En | MEDLINE | ID: mdl-29875417

SETBP1 variants occur as somatic mutations in several hematological malignancies such as atypical chronic myeloid leukemia and as de novo germline mutations in the Schinzel-Giedion syndrome. Here we show that SETBP1 binds to gDNA in AT-rich promoter regions, causing activation of gene expression through recruitment of a HCF1/KMT2A/PHF8 epigenetic complex. Deletion of two AT-hooks abrogates the binding of SETBP1 to gDNA and impairs target gene upregulation. Genes controlled by SETBP1 such as MECOM are significantly upregulated in leukemias containing SETBP1 mutations. Gene ontology analysis of deregulated SETBP1 target genes indicates that they are also key controllers of visceral organ development and brain morphogenesis. In line with these findings, in utero brain electroporation of mutated SETBP1 causes impairment of mouse neurogenesis with a profound delay in neuronal migration. In summary, this work unveils a SETBP1 function that directly affects gene transcription and clarifies the mechanism operating in myeloid malignancies and in the Schinzel-Giedion syndrome caused by SETBP1 mutations.


Carrier Proteins/genetics , Epigenesis, Genetic , Gene Expression Profiling , Mutation , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Abnormalities, Multiple/genetics , Animals , Brain/embryology , Brain/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Craniofacial Abnormalities/genetics , Gene Ontology , HEK293 Cells , Hand Deformities, Congenital/genetics , Humans , Intellectual Disability/genetics , Leukemia/genetics , Leukemia/pathology , Mice , Nails, Malformed/genetics , Neurogenesis/genetics , Nuclear Proteins/metabolism , Protein Binding
17.
Dev Biol ; 434(2): 231-248, 2018 02 15.
Article En | MEDLINE | ID: mdl-29305158

During cerebral cortex development, neural progenitors are required to elaborate a variety of cell differentiation signals to which they are continuously exposed. RA acid is a potent inducer of neuronal differentiation as it was found to influence cortical development. We report herein that TBR2, a transcription factor specific to Intermediate (Basal) Neural Progenitors (INPs), represses activation of the RA responsive element and expression of RA target genes in cell lines. This repressive action on RA signaling was functionally confirmed by the decrease of RA-mediated neuronal differentiation in neural stem cells stably overexpressing TBR2. In vivo mapping of RA activity in the developing cortex indicated that RA activity is detected in radial glial cells and subsequently downregulated in INPs, revealing a fine cell-type specific regulation of its signaling. Thus, TBR2 might be a molecular player in opposing RA signaling in INPs. Interestingly, this negative regulation is achieved at least in part by directly repressing the critical nuclear RA co-factor ZFP423. Indeed, we found ZFP423 to be expressed in the developing cortex and promote RA-dependent neuronal differentiation. These data indicate that TBR2 contributes to suppressing RA signaling in INPs, thereby enabling them to re-enter the cell cycle and delay neuronal differentiation.


Cell Differentiation/drug effects , Cerebral Cortex/embryology , DNA-Binding Proteins/metabolism , Neural Stem Cells/metabolism , Organogenesis/drug effects , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cerebral Cortex/cytology , DNA-Binding Proteins/genetics , Mice , Neural Stem Cells/cytology , Organogenesis/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , T-Box Domain Proteins/genetics , Transcription Factors/genetics
18.
J Cell Physiol ; 233(4): 3152-3163, 2018 04.
Article En | MEDLINE | ID: mdl-28816361

mSEL-1L is a highly conserved ER-resident type I protein, involved in the degradation of misfolded peptides through the ubiquitin-proteasome system (UPS), a pathway known to control the plasticity of the vascular smooth muscle cells (VSMC) phenotype and survival. In this article, we demonstrate that mSEL-1L deficiency interferes with the murine embryonic vascular network, showing particular irregularities in the intracranic and intersomitic neurovascular units and in the cerebral capillary microcirculation. During murine embryogenesis, mSEL-1L is expressed in cerebral areas known to harbor progenitor neural cells, while in the adult brain the protein is specifically restricted to the stem cell niches, co-localizing with Sox2 and Nestin. Null mice are characterized by important defects in the development of telenchephalic regions, revealing conspicuous aberration in neural stem cell lineage commitment. Moreover, mSEL-1L depletion in vitro and in vivo appears to affect the harmonic differentiation of the NSCs, by negatively influencing the corticogenesis processes. Overall, the data presented suggests that the drastic phenotypic characteristics exhibited in mSEL-1L null mice can, in part, be explained by the negative influence it plays on Notch1 signaling pathway.


Cell Lineage , Neovascularization, Physiologic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Proteins/metabolism , Animals , Brain/growth & development , Brain/metabolism , Cell Proliferation , Cell Self Renewal , Genome , Intracellular Signaling Peptides and Proteins , Mice, Knockout , Receptors, Notch/metabolism , Transcriptome/genetics
20.
Mol Ther ; 25(12): 2727-2742, 2017 Dec 06.
Article En | MEDLINE | ID: mdl-28882452

The lack of technology for direct global-scale targeting of the adult mouse nervous system has hindered research on brain processing and dysfunctions. Currently, gene transfer is normally achieved by intraparenchymal viral injections, but these injections target a restricted brain area. Herein, we demonstrated that intravenous delivery of adeno-associated virus (AAV)-PHP.B viral particles permeated and diffused throughout the neural parenchyma, targeting both the central and the peripheral nervous system in a global pattern. We then established multiple procedures of viral transduction to control gene expression or inactivate gene function exclusively in the adult nervous system and assessed the underlying behavioral effects. Building on these results, we established an effective gene therapy strategy to counteract the widespread accumulation of α-synuclein deposits throughout the forebrain in a mouse model of synucleinopathy. Transduction of A53T-SCNA transgenic mice with AAV-PHP.B-GBA1 restored physiological levels of the enzyme, reduced α-synuclein pathology, and produced significant behavioral recovery. Finally, we provided evidence that AAV-PHP.B brain penetration does not lead to evident dysfunctions in blood-brain barrier integrity or permeability. Altogether, the AAV-PHP.B viral platform enables non-invasive, widespread, and long-lasting global neural expression of therapeutic genes, such as GBA1, providing an invaluable approach to treat neurodegenerative diseases with diffuse brain pathology such as synucleinopathies.


Dependovirus/genetics , Gene Expression , Genetic Vectors/genetics , beta-Glucosidase/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Disease Models, Animal , Electroencephalography , Enzyme Activation , Gene Order , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Transduction, Genetic , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
...