Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Cell Mol Life Sci ; 79(1): 56, 2021 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-34921638

RESUMO

The human cerebral cortex is a uniquely complex structure encompassing an unparalleled diversity of neuronal types and subtypes. These arise during development through a series of evolutionary conserved processes, such as progenitor cell proliferation, migration and differentiation, incorporating human-associated adaptations including a protracted neurogenesis and the emergence of novel highly heterogeneous progenitor populations. Disentangling the unique features of human cortical development involves elucidation of the intricate developmental cell transitions orchestrated by progressive molecular events. Crucially, developmental timing controls the fine balance between cell cycle progression/exit and the neurogenic competence of precursor cells, which undergo morphological transitions coupled to transcriptome-defined temporal states. Recent advances in bulk and single-cell transcriptomic technologies suggest that alongside protein-coding genes, non-coding RNAs exert important regulatory roles in these processes. Interestingly, a considerable number of novel long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have appeared in human and non-human primates suggesting an evolutionary role in shaping cortical development. Here, we present an overview of human cortical development and highlight the marked diversification and complexity of human neuronal progenitors. We further discuss how lncRNAs and miRNAs constitute critical components of the extended epigenetic regulatory network defining intermediate states of progenitors and controlling cell cycle dynamics and fate choices with spatiotemporal precision, during human neurodevelopment.


Assuntos
Córtex Cerebral , MicroRNAs/fisiologia , Neurogênese , Neurônios/metabolismo , RNA Longo não Codificante/fisiologia , Diferenciação Celular , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Humanos , Neurônios/citologia
2.
PLoS Genet ; 13(4): e1006656, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28376086

RESUMO

The mitochondrial protein SLC25A46 has been recently identified as a novel pathogenic cause in a wide spectrum of neurological diseases, including inherited optic atrophy, Charcot-Marie-Tooth type 2, Leigh syndrome, progressive myoclonic ataxia and lethal congenital pontocerebellar hypoplasia. SLC25A46 is an outer membrane protein, member of the Solute Carrier 25 (SLC25) family of nuclear genes encoding mitochondrial carriers, with a role in mitochondrial dynamics and cristae maintenance. Here we identified a loss-of-function mutation in the Slc25a46 gene that causes lethal neuropathology in mice. Mutant mice manifest the main clinical features identified in patients, including ataxia, optic atrophy and cerebellar hypoplasia, which were completely rescued by expression of the human ortholog. Histopathological analysis revealed previously unseen lesions, most notably disrupted cytoarchitecture in the cerebellum and retina and prominent abnormalities in the neuromuscular junction. A distinct lymphoid phenotype was also evident. Our mutant mice provide a valid model for understanding the mechanistic basis of the complex SLC25A46-mediated pathologies, as well as for screening potential therapeutic interventions.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Mutação/genética , Proteínas de Transporte de Fosfato/genética , Animais , Ataxia/genética , Ataxia/fisiopatologia , Doenças Cerebelares/genética , Doenças Cerebelares/fisiopatologia , Doença de Charcot-Marie-Tooth/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Membranas Mitocondriais/metabolismo , Atrofia Óptica/genética , Atrofia Óptica/fisiopatologia , Linhagem , Fenótipo
3.
Proc Natl Acad Sci U S A ; 114(18): E3679-E3688, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28416701

RESUMO

α-Synuclein (αSyn) is the major gene linked to sporadic Parkinson's disease (PD), whereas the G209A (p.A53T) αSyn mutation causes a familial form of PD characterized by early onset and a generally severe phenotype, including nonmotor manifestations. Here we generated de novo induced pluripotent stem cells (iPSCs) from patients harboring the p.A53T mutation and developed a robust model that captures PD pathogenic processes under basal conditions. iPSC-derived mutant neurons displayed novel disease-relevant phenotypes, including protein aggregation, compromised neuritic outgrowth, and contorted or fragmented axons with swollen varicosities containing αSyn and Tau. The identified neuropathological features closely resembled those in brains of p.A53T patients. Small molecules targeting αSyn reverted the degenerative phenotype under both basal and induced stress conditions, indicating a treatment strategy for PD and other synucleinopathies. Furthermore, mutant neurons showed disrupted synaptic connectivity and widespread transcriptional alterations in genes involved in synaptic signaling, a number of which have been previously linked to mental disorders, raising intriguing implications for potentially converging disease mechanisms.


Assuntos
Axônios/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Mutação de Sentido Incorreto , Doença de Parkinson/metabolismo , Polineuropatias/metabolismo , Transmissão Sináptica , alfa-Sinucleína/metabolismo , Substituição de Aminoácidos , Axônios/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Polineuropatias/genética , Polineuropatias/patologia , alfa-Sinucleína/genética
4.
Int J Mol Sci ; 21(19)2020 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993172

RESUMO

Parkinson's disease (PD) is a common progressive neurodegenerative disorder characterized by loss of striatal-projecting dopaminergic neurons of the ventral forebrain, resulting in motor and cognitive deficits. Despite extensive efforts in understanding PD pathogenesis, no disease-modifying drugs exist. Recent advances in cell reprogramming technologies have facilitated the generation of patient-derived models for sporadic or familial PD and the identification of early, potentially triggering, pathological phenotypes while they provide amenable systems for drug discovery. Emerging developments highlight the enhanced potential of using more sophisticated cellular systems, including neuronal and glial co-cultures as well as three-dimensional systems that better simulate the human pathophysiology. In combination with high-throughput high-content screening technologies, these approaches open new perspectives for the identification of disease-modifying compounds. In this review, we discuss current advances and the challenges ahead in the use of patient-derived induced pluripotent stem cells for drug discovery in PD. We address new concepts implicating non-neuronal cells in disease pathogenesis and highlight the necessity for functional assays, such as calcium imaging and multi-electrode array recordings, to predict drug efficacy. Finally, we argue that artificial intelligence technologies will be pivotal for analysis of the large and complex data sets obtained, becoming game-changers in the process of drug discovery.


Assuntos
Descoberta de Drogas/métodos , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios/patologia , Doença de Parkinson/patologia , Animais , Técnicas de Cocultura/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico
5.
Neurobiol Dis ; 132: 104609, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31494284

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder whereby loss of midbrain dopaminergic neurons results in motor dysfunction. Transplantation of human induced pluripotent stem cells (iPSCs) into the brain of patients affected by PD is one of the therapeutic approaches that has gained interest to compensate for the degeneration of neurons and improve disease symptoms. However, only a part of transplanted cells can differentiate into mature neurons while the majority remains in undifferentiated state. Here we investigated whether human neuronal precursor cells (hNPCs) derived from iPSCs have an active role in α-synuclein (α-syn) pathology. Our findings demonstrate that α-syn fibrils are taken up by hNPCs and are preferentially localized in lysosomes where they can be degraded. However, α-syn fibrils are also transferred between hNPCs in a cell-to-cell contact dependent manner, and are found in tunneling nanotube (TNT)-like structures. Thus, NPCs can have a dual role in the progression of α-syn pathology, which should be considered in human transplants.


Assuntos
Comunicação Celular/fisiologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/ultraestrutura , Endocitose/fisiologia , alfa-Sinucleína/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Lisossomos/metabolismo , Células-Tronco Neurais/metabolismo
6.
Neurochem Res ; 44(6): 1475-1493, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30989481

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder. We have previously developed a disease-in-a-dish model for familial PD using induced pluripotent stem cells (iPSCs) from two patients carrying the p.A53T α-synuclein (αSyn) mutation. By directed differentiation, we generated a model that displays disease-relevant phenotypes, including protein aggregation, compromised neurite outgrowth, axonal neuropathology and synaptic defects. Here we investigated the in vivo phenotypes of iPSCs, derived from one patient, after transplantation in a lesion mouse model established by unilateral intrastriatal 6-hydroxydopamine injection in the immunosuppressed NOD/SCID strain. Immunohistochemistry revealed that despite the disease-related characteristics that mutant cells displayed when maintained up to 70 days in vitro, they could survive and differentiate in vivo over a 12-week period. However, some differences were noted between patient-derived and control grafts, including a significant rise in αSyn immunoreactivity that might signal a first step towards pathology. Moreover, control-derived grafts appeared to integrate better than PD grafts within the host tissue extending projections that formed more contacts with host striatal neurons. Our data suggest that the distinct disease-related characteristics which p.A53T cells develop in vitro, may be attenuated or take longer to emerge in vivo after transplantation within the mouse brain. Further analysis of the phenotypes that patient cells acquire over longer periods of time as well as the use of multiple iPSC clones from different patients should extend our current proof-of-concept study and provide additional evidence for in vivo disease modeling.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Doença de Parkinson , Fenótipo , Animais , Encéfalo/citologia , Encéfalo/cirurgia , Neurônios Dopaminérgicos/citologia , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação , Estudo de Prova de Conceito , Transplante Heterólogo , alfa-Sinucleína/genética
7.
Glia ; 64(5): 763-79, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26712314

RESUMO

The central nervous system has limited capacity for regeneration after traumatic injury. Transplantation of neural stem/progenitor cells (NPCs) has been proposed as a potential therapeutic approach while insulin-like growth factor I (IGF-I) has neuroprotective properties following various experimental insults to the nervous system. We have previously shown that NPCs transduced with a lentiviral vector for IGF-I overexpression have an enhanced ability to give rise to neurons in vitro but also in vivo, upon transplantation in a mouse model of temporal lobe epilepsy. Here we studied the regenerative potential of NPCs, IGF-I-transduced or not, in a mouse model of hippocampal mechanical injury. NPC transplantation, with or without IGF-I transduction, rescued the injury-induced spatial learning deficits as revealed in the Morris Water Maze. Moreover, it had beneficial effects on the host tissue by reducing astroglial activation and microglial/macrophage accumulation while enhancing generation of endogenous oligodendrocyte precursor cells. One or two months after transplantation the grafted NPCs had migrated towards the lesion site and in the neighboring myelin-rich regions. Transplanted cells differentiated toward the oligodendroglial, but not the neuronal or astrocytic lineages, expressing the early and late oligodendrocyte markers NG2, Olig2, and CNPase. The newly generated oligodendrocytes reached maturity and formed myelin internodes. Our current and previous observations illustrate the high plasticity of transplanted NPCs which can acquire injury-dependent phenotypes within the host CNS, supporting the fact that reciprocal interactions between transplanted cells and the host tissue are an important factor to be considered when designing prospective cell-based therapies for CNS degenerative conditions.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/cirurgia , Diferenciação Celular/fisiologia , Inflamação/etiologia , Deficiências da Aprendizagem/etiologia , Oligodendroglia/fisiologia , Transplante de Células-Tronco/métodos , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Antígenos CD/metabolismo , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/cirurgia , Antígeno Ki-67/metabolismo , Deficiências da Aprendizagem/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Proteoglicanas/metabolismo
8.
Mol Ther ; 23(6): 993-1002, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25807288

RESUMO

Failure of the mammalian central nervous system (CNS) to regenerate effectively after injury leads to mostly irreversible functional impairment. Gold nanoparticles (AuNPs) are promising candidates for drug delivery in combination with tissue-compatible reagents, such as polyethylene glycol (PEG). PEG administration in CNS injury models has received interest for potential therapy, but toxicity and low bioavailability prevents clinical application. Here we show that intraspinal delivery of PEG-functionalized 40-nm-AuNPs at early stages after mouse spinal cord injury is beneficial for recovery. Positive outcome of hind limb motor function was accompanied by attenuated inflammatory response, enhanced motor neuron survival, and increased myelination of spared or regrown/sprouted axons. No adverse effects, such as body weight loss, ill health, or increased mortality were observed. We propose that PEG-AuNPs represent a favorable drug-delivery platform with therapeutic potential that could be further enhanced if PEG-AuNPs are used as carriers of regeneration-promoting molecules.


Assuntos
Materiais Revestidos Biocompatíveis/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Ouro/farmacologia , Nanopartículas Metálicas/química , Polietilenoglicóis/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Materiais Revestidos Biocompatíveis/química , Modelos Animais de Doenças , Feminino , Ouro/química , Camundongos , Camundongos Endogâmicos C57BL , Polietilenoglicóis/química , Recuperação de Função Fisiológica/efeitos dos fármacos
9.
Glia ; 63(7): 1101-25, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25731941

RESUMO

Myelin integrity is crucial for central nervous system (CNS) physiology while its preservation and regeneration after spinal cord injury (SCI) is key to functional restoration. Disturbance of nodal organization acutely after SCI exposes the axon and triggers conduction block in the absence of overt demyelination. Oligodendrocyte (OL) loss and myelin degradation follow as a consequence of secondary damage. Here, we provide an overview of the major biological events and underlying mechanisms leading to OL death and demyelination and discuss strategies to restrain these processes. Another aspect which is critical for SCI repair is the enhancement of endogenously occurring spontaneous remyelination. Recent findings have unveiled the complex roles of innate and adaptive immune responses in remyelination and the immunoregulatory potential of the glial scar. Moreover, the intimate crosstalk between neuronal activity, oligodendrogenesis and myelination emphasizes the contribution of rehabilitation to functional recovery. With a view toward clinical applications, several therapeutic strategies have been devised to target SCI pathology, including genetic manipulation, administration of small therapeutic molecules, immunomodulation, manipulation of the glial scar and cell transplantation. The implementation of new tools such as cellular reprogramming for conversion of one somatic cell type to another or the use of nanotechnology and tissue engineering products provides additional opportunities for SCI repair. Given the complexity of the spinal cord tissue after injury, it is becoming apparent that combinatorial strategies are needed to rescue OLs and myelin at early stages after SCI and support remyelination, paving the way toward clinical translation.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Doenças Desmielinizantes/terapia , Bainha de Mielina/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia , Regeneração da Medula Espinal/fisiologia , Animais , Humanos , Oligodendroglia
10.
Stem Cells ; 32(6): 1674-87, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24497115

RESUMO

Cellular prion protein (PrP) is prominently expressed in brain, in differentiated neurons but also in neural stem/precursor cells (NPCs). The misfolding of PrP is a central event in prion diseases, yet the physiological function of PrP is insufficiently understood. Although PrP has been reported to associate with the neural cell adhesion molecule (NCAM), the consequences of concerted PrP-NCAM action in NPC physiology are unknown. Here, we generated NPCs from the subventricular zone (SVZ) of postnatal day 5 wild-type and PrP null (-/-) mice and observed that PrP is essential for proper NPC proliferation and neuronal differentiation. Moreover, we found that PrP is required for the NPC response to NCAM-induced neuronal differentiation. In the absence of PrP, NCAM not only fails to promote neuronal differentiation but also induces an accumulation of doublecortin-positive neuronal progenitors at the proliferation stage. In agreement, we noted an increase in cycling neuronal progenitors in the SVZ of PrP-/- mice compared with PrP+/+ mice, as evidenced by double labeling for the proliferation marker Ki67 and doublecortin as well as by 5-bromo-2'-deoxyuridine incorporation experiments. Additionally, fewer newly born neurons were detected in the rostral migratory stream of PrP-/- mice. Analysis of the migration of SVZ cells in microexplant cultures from wild-type and PrP-/- mice revealed no differences between genotypes or a role for NCAM in this process. Our data demonstrate that PrP plays a critical role in neuronal differentiation of NPCs and suggest that this function is, at least in part, NCAM-dependent.


Assuntos
Diferenciação Celular , Moléculas de Adesão de Célula Nervosa/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Príons/metabolismo , Animais , Bromodesoxiuridina/metabolismo , Ciclo Celular , Movimento Celular , Proliferação de Células , Ventrículos Cerebrais/citologia , Proteínas do Domínio Duplacortina , Células HEK293 , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo
11.
PLoS Biol ; 8(12): e1000565, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21203589

RESUMO

Activation of Notch1 signaling in neural progenitor cells (NPCs) induces self-renewal and inhibits neurogenesis. Upon neuronal differentiation, NPCs overcome this inhibition, express proneural genes to induce Notch ligands, and activate Notch1 in neighboring NPCs. The molecular mechanism that coordinates Notch1 inactivation with initiation of neurogenesis remains elusive. Here, we provide evidence that Prox1, a transcription repressor and downstream target of proneural genes, counteracts Notch1 signaling via direct suppression of Notch1 gene expression. By expression studies in the developing spinal cord of chick and mouse embryo, we showed that Prox1 is limited to neuronal precursors residing between the Notch1+ NPCs and post-mitotic neurons. Physiological levels of Prox1 in this tissue are sufficient to allow binding at Notch1 promoter and they are critical for proper Notch1 transcriptional regulation in vivo. Gain-of-function studies in the chick neural tube and mouse NPCs suggest that Prox1-mediated suppression of Notch1 relieves its inhibition on neurogenesis and allows NPCs to exit the cell cycle and differentiate. Moreover, loss-of-function in the chick neural tube shows that Prox1 is necessary for suppression of Notch1 outside the ventricular zone, inhibition of active Notch signaling, down-regulation of NPC markers, and completion of neuronal differentiation program. Together these data suggest that Prox1 inhibits Notch1 gene expression to control the balance between NPC self-renewal and neuronal differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/metabolismo , Tubo Neural/metabolismo , Receptor Notch1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular , Embrião de Galinha , Proteínas de Homeodomínio/genética , Camundongos , Células-Tronco Neurais/citologia , Tubo Neural/citologia , Tubo Neural/embriologia , Neurogênese , Receptor Notch1/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
12.
Cells ; 12(12)2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37371040

RESUMO

Central nervous system infections caused by pathogens crossing the blood-brain barrier are extremely damaging and trigger cellular alterations and neuroinflammation. Bacterial brain infection, in particular, is a major cause of hippocampal neuronal degeneration. Hippocampal neurogenesis, a continuous multistep process occurring throughout life in the adult brain, could compensate for such neuronal loss. However, the high rates of cognitive and other sequelae from bacterial meningitis/encephalitis suggest that endogenous repair mechanisms might be severely affected. In the current study, we used Group B Streptococcus (GBS) strain NEM316, to establish an adult mouse model of brain infection and determine its impact on adult neurogenesis. Experimental encephalitis elicited neurological deficits and death, induced inflammation, and affected neurogenesis in the dentate gyrus of the adult hippocampus by suppressing the proliferation of progenitor cells and the generation of newborn neurons. These effects were specifically associated with hippocampal neurogenesis while subventricular zone neurogenesis was not affected. Overall, our data provide new insights regarding the effect of GBS infection on adult brain neurogenesis.


Assuntos
Encefalite , Neurogênese , Camundongos , Animais , Neurogênese/fisiologia , Hipocampo , Inflamação , Streptococcus
13.
Int J Dev Biol ; 66(1-2-3): 23-33, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34881786

RESUMO

Stem cell technologies have opened up new avenues in the study of human biology and disease. In particular, the advent of human embryonic stem cells followed by reprograming technologies for generation of induced pluripotent stem cells have instigated studies into modeling human brain development and disease by providing a means to simulate a human tissue otherwise completely or largely inaccessible to researchers. Brain development is a complex process achieved in a remarkably controlled spatial and temporal manner through coordinated cellular and molecular events. In vitro models aim to mimic these processes and recapitulate brain organogenesis. Initially, two-dimensional neural cultures presented an innovative landmark for investigating human neuronal and, more recently, glial biology, as well as for modeling brain neurodevelopmental and neurodegenerative diseases. The establishment of three-dimensional cultures in the form of brain organoids was an equally important milestone in the field. Brain organoids mimic more closely the in vivo tissue composition and architecture and are more physiologically relevant than monolayer cultures. They therefore represent a more realistic cellular environment for modeling the cell biology and pathology of the nervous system. Here we highlight the journey towards recapitulating human brain development and disease in a dish, progressing from two-dimensional in vitro systems to the third dimension provided by brain organoids. We discuss the potential of these approaches for modeling human brain development and evolution, and their promising contribution towards understanding and treating brain disease.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Encéfalo/fisiologia , Humanos , Organogênese/fisiologia , Organoides/fisiologia
14.
Biomolecules ; 12(7)2022 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-35883433

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder, classically associated with extensive loss of dopaminergic neurons of the substantia nigra pars compacta. The hallmark of the disease is the accumulation of pathogenic conformations of the presynaptic protein, α-synuclein (αSyn), and the formation of intraneuronal protein aggregate inclusions. Neurodegeneration of dopamine neurons leads to a prominent dopaminergic deficiency in the basal ganglia, responsible for motor disturbances. However, it is now recognized that the disease involves more widespread neuronal dysfunction, leading to early and late non-motor symptoms. The development of in vitro systems based on the differentiation of human-induced pluripotent stem cells provides us the unique opportunity to monitor alterations at the cellular and molecular level throughout the differentiation procedure and identify perturbations that occur early, even at the neuronal precursor stage. Here we aim to identify whether p.A53T-αSyn induced disturbances at the molecular level are already present in neural precursors. Towards this, we present data from transcriptomics analysis of control and p.A53T-αSyn NPCs showing altered expression in transcripts involved in axon guidance, adhesion, synaptogenesis, ion transport, and metabolism. The comparative analysis with the transcriptomics profile of p.A53T-αSyn neurons shows both distinct and overlapping pathways leading to neurodegeneration while meta-analysis with transcriptomics data from both neurodegenerative and neurodevelopmental disorders reveals that p.A53T-pathology has a significant overlap with the latter category. This is the first study showing that molecular dysregulation initiates early at the p.A53T-αSyn NPC level, suggesting that synucleinopathies may have a neurodevelopmental component.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Sinucleinopatias , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
NPJ Parkinsons Dis ; 8(1): 15, 2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149677

RESUMO

Combining high throughput screening approaches with induced pluripotent stem cell (iPSC)-based disease modeling represents a promising unbiased strategy to identify therapies for neurodegenerative disorders. Here we applied high content imaging on iPSC-derived neurons from patients with familial Parkinson's disease bearing the G209A (p.A53T) α-synuclein (αSyn) mutation and launched a screening campaign on a small kinase inhibitor library. We thus identified the multi-kinase inhibitor BX795 that at a single dose effectively restores disease-associated neurodegenerative phenotypes. Proteomics profiling mapped the molecular pathways underlying the protective effects of BX795, comprising a cohort of 118 protein-mediators of the core biological processes of RNA metabolism, protein synthesis, modification and clearance, and stress response, all linked to the mTORC1 signaling hub. In agreement, expression of human p.A53T-αSyn in neuronal cells affected key components of the mTORC1 pathway resulting in aberrant protein synthesis that was restored in the presence of BX795 with concurrent facilitation of autophagy. Taken together, we have identified a promising small molecule with neuroprotective actions as candidate therapeutic for PD and other protein conformational disorders.

16.
Stem Cells ; 28(1): 127-39, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19911428

RESUMO

The intrinsic inability of the central nervous system to efficiently repair traumatic injuries renders transplantation of neural stem/precursor cells (NPCs) a promising approach towards repair of brain lesions. In this study, NPCs derived from embryonic day 14.5 mouse cortex were genetically modified via transduction with a lentiviral vector to overexpress the neuronal lineage-specific regulator BM88/Cend1 that coordinates cell cycle exit and differentiation of neuronal precursors. BM88/Cend1-overexpressing NPCs exhibiting enhanced differentiation into neurons in vitro were transplanted in a mouse model of acute cortical injury and analyzed in comparison with control NPCs. Immunohistochemical analysis revealed that a smaller proportion of BM88/Cend1-overexpressing NPCs, as compared with control NPCs, expressed the neural stem cell marker nestin 1 day after transplantation, while the percentage of nestin-positive cells was significantly reduced thereafter in both types of cells, being almost extinct 1 week post-grafting. Both types of cells did not proliferate up to 4 weeks in vivo, thus minimizing the risk of tumorigenesis. In comparison with control NPCs, Cend1-overexpressing NPCs generated more neurons and less glial cells 1 month after transplantation in the lesioned cortex whereas the majority of graft-derived neurons were identified as GABAergic interneurons. Furthermore, transplantation of Cend1-overexpressing NPCs resulted in a marked reduction of astrogliosis around the lesioned area as compared to grafts of control NPCs. Our results suggest that transplantation of Cend1-overexpressing NPCs exerts beneficial effects on tissue regeneration by enhancing the number of generated neurons and restricting the formation of astroglial scar, in a mouse model of cortical brain injury.


Assuntos
Lesões Encefálicas/terapia , Células-Tronco Embrionárias/transplante , Terapia Genética , Proteínas de Membrana/biossíntese , Córtex Motor/fisiopatologia , Regeneração Nervosa , Proteínas do Tecido Nervoso/biossíntese , Neurônios/transplante , Transplante de Células-Tronco , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células-Tronco Embrionárias/metabolismo , Feminino , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/metabolismo , Lentivirus/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Córtex Motor/embriologia , Córtex Motor/lesões , Córtex Motor/metabolismo , Córtex Motor/patologia , Destreza Motora , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Recuperação de Função Fisiológica , Esferoides Celulares , Suínos , Fatores de Tempo , Transdução Genética
17.
Mol Cell Neurosci ; 44(1): 15-29, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20153830

RESUMO

During nervous system development, neural progenitors arise in proliferative zones, then exit the cell cycle and differentiate as they migrate away from these zones. The neuronal protein BM88/Cend1 has been implicated in coordination of cell cycle exit and differentiation of neuronal precursors. To further elucidate its function we generated Cend1 knock-out mice and analyzed their phenotype during postnatal cerebellar development. Cend1(-/-) mice showed no overt abnormalities in the gross anatomy of the cerebellum or other brain regions. However, detailed analysis revealed alterations in cerebellar layering arising from increased proliferation of granule cell precursors, delayed radial granule cell migration and impaired Purkinje cell differentiation. Accordingly, expression of Patched1, cyclin D1, reelin and brain-derived neurotrophic factor, which correlate with morphological development of the cerebellum, was altered in Cend1(-/-) mice. The observed anatomical and molecular alterations were accompanied by deficits in motor behaviour. Our results suggest that Cend1 is required for normal cerebellar development.


Assuntos
Cerebelo/anormalidades , Cerebelo/metabolismo , Proteínas de Membrana/genética , Transtornos dos Movimentos/genética , Proteínas do Tecido Nervoso/genética , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Diferenciação Celular/genética , Movimento Celular/genética , Forma Celular/genética , Células Cultivadas , Cerebelo/patologia , Dendritos/metabolismo , Dendritos/patologia , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Citometria por Imagem , Camundongos , Camundongos Knockout , Transtornos dos Movimentos/patologia , Transtornos dos Movimentos/fisiopatologia , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/metabolismo , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Neurogênese/genética , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Proteína Reelina , Coloração pela Prata
18.
Front Microbiol ; 12: 730892, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970230

RESUMO

Rabies is a zoonotic disease caused by rabies virus (RABV). As rabies advances, patients develop a variety of severe neurological symptoms that inevitably lead to coma and death. Unlike other neurotropic viruses that can induce symptoms of a similar range, RABV-infected post-mortem brains do not show significant signs of inflammation nor the structural damages on neurons. This suggests that the observed neurological symptoms possibly originate from dysfunctions of neurons. However, many aspects of neuronal dysfunctions in the context of RABV infection are only partially understood, and therefore require further investigation. In this study, we used differentiated neurons to characterize the RABV-induced transcriptomic changes at the early time-points of infection. We found that the genes modulated in response to the infection are particularly involved in cell cycle, gene expression, immune response, and neuronal function-associated processes. Comparing a wild-type RABV to a mutant virus harboring altered matrix proteins, we found that the RABV matrix protein plays an important role in the early down-regulation of host genes, of which a significant number is involved in neuronal functions. The kinetics of differentially expressed genes (DEGs) are also different between the wild type and mutant virus datasets. The number of modulated genes remained constant upon wild-type RABV infection up to 24 h post-infection, but dramatically increased in the mutant condition. This result suggests that the intact viral matrix protein is important to control the size of host gene modulation. We then examined the signaling pathways previously studied in relation to the innate immune responses against RABV, and found that these pathways contribute to the changes in neuronal function-associated processes. We further examined a set of regulated genes that could impact neuronal functions collectively, and demonstrated in calcium imaging that indeed the spontaneous activity of neurons is influenced by RABV infection. Overall, our findings suggest that neuronal function-associated genes are modulated by RABV early on, potentially through the viral matrix protein-interacting signaling molecules and their downstream pathways.

19.
Free Radic Biol Med ; 162: 88-103, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33279620

RESUMO

Alzheimer's disease (AD) is the most common form of dementia worldwide, characterized by a progressive decline in a variety of cognitive and non-cognitive functions. The amyloid beta protein cascade hypothesis places the formation of amyloid beta protein aggregates on the first position in the complex pathological cascade leading to neurodegeneration, and therefore AD might be considered to be a protein-misfolding disease. The Ubiquitin Proteasome System (UPS), being the primary protein degradation mechanism with a fundamental role in the maintenance of proteostasis, has been identified as a putative therapeutic target to delay and/or to decelerate the progression of neurodegenerative disorders that are characterized by accumulated/aggregated proteins. The purpose of this study was to test if the activation of proteasome in vivo can alleviate AD pathology. Specifically by using two compounds with complementary modes of proteasome activation and documented antioxidant and redox regulating properties in the 5xFAD transgenic mice model of AD, we ameliorated a number of AD related deficits. Shortly after proteasome activation we detected significantly reduced amyloid-beta load correlated with improved motor functions, reduced anxiety and frailty level. Essentially, to our knowledge this is the first report to demonstrate a dual activation of the proteasome and its downstream effects. In conclusion, these findings open up new directions for future therapeutic potential of proteasome-mediated proteolysis enhancement.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Fenótipo , Complexo de Endopeptidases do Proteassoma
20.
J Neurochem ; 115(2): 460-74, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20681949

RESUMO

Strategies to enhance neural stem/precursor cell (NPC) capacity to yield multipotential, proliferative, and migrating pools of cells that can efficiently differentiate into neurons could be crucial for structural repair after neurodegenerative damage. Here, we have generated a lentiviral vector for expression of insulin-like growth factor-I (IGF-1) and investigated the impact of IGF-1 transduction on the properties of cultured NPCs (IGF-1-NPCs). Under proliferative conditions, IGF-1 transduction promoted cell cycle progression via cyclin D1 up-regulation and Akt phosphorylation. Remarkably upon differentiation-inducing conditions, IGF-1-NPCs cease to proliferate and differentiate to a greater extent into neurons with significantly longer neurites, at the expense of astrocytes. Moreover, using live imaging we provide evidence that IGF-1 transduction enhances the motility and tissue penetration of grafted NPCs in cultured cortical slices. These results illustrate the important consequence of IGF-1 transduction in regulating NPC functions and offer a potential strategy to enhance the prospective repair potential of NPCs.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Fator de Crescimento Insulin-Like I/metabolismo , Neurônios/fisiologia , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Diferenciação Celular/genética , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ventrículos Cerebrais/citologia , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Citometria de Fluxo/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Hidroxiureia/farmacologia , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/farmacologia , Lentivirus/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Transdução Genética/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA