Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-36645345

RESUMO

Wolfram syndrome 1 (WS1) is a rare genetic disorder caused by mutations in the WFS1 gene leading to a wide spectrum of clinical dysfunctions, among which blindness, diabetes, and neurological deficits are the most prominent. WFS1 encodes for the endoplasmic reticulum (ER) resident transmembrane protein wolframin with multiple functions in ER processes. However, the WFS1-dependent etiopathology in retinal cells is unknown. Herein, we showed that Wfs1 mutant mice developed early retinal electrophysiological impairments followed by marked visual loss. Interestingly, axons and myelin disruption in the optic nerve preceded the degeneration of the retinal ganglion cell bodies in the retina. Transcriptomics at pre-degenerative stage revealed the STAT3-dependent activation of proinflammatory glial markers with reduction of the homeostatic and pro-survival factors glutamine synthetase and BDNF. Furthermore, label-free comparative proteomics identified a significant reduction of the monocarboxylate transport isoform 1 (MCT1) and its partner basigin that are highly enriched on retinal glia and myelin-forming oligodendrocytes in optic nerve together with wolframin. Loss of MCT1 caused a failure in lactate transfer from glial to neuronal cell bodies and axons leading to a chronic hypometabolic state. Thus, this bioenergetic impairment is occurring concurrently both within the axonal regions and cell bodies of the retinal ganglion cells, selectively endangering their survival while impacting less on other retinal cells. This metabolic dysfunction occurs months before the frank RGC degeneration suggesting an extended time-window for intervening with new therapeutic strategies focused on boosting retinal and optic nerve bioenergetics in WS1.


Assuntos
Atrofia Óptica , Síndrome de Wolfram , Animais , Camundongos , Degeneração Neural/metabolismo , Doenças Neuroinflamatórias , Células Ganglionares da Retina/metabolismo , Síndrome de Wolfram/genética , Síndrome de Wolfram/metabolismo
2.
Hum Mol Genet ; 27(5): 761-779, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29281027

RESUMO

P23H is the most common mutation in the RHODOPSIN (RHO) gene leading to a dominant form of retinitis pigmentosa (RP), a rod photoreceptor degeneration that invariably causes vision loss. Specific disruption of the disease P23H RHO mutant while preserving the wild-type (WT) functional allele would be an invaluable therapy for this disease. However, various technologies tested in the past failed to achieve effective changes and consequently therapeutic benefits. We validated a CRISPR/Cas9 strategy to specifically inactivate the P23H RHO mutant, while preserving the WT allele in vitro. We, then, translated this approach in vivo by delivering the CRISPR/Cas9 components in murine Rho+/P23H mutant retinae. Targeted retinae presented a high rate of cleavage in the P23H but not WT Rho allele. This gene manipulation was sufficient to slow photoreceptor degeneration and improve retinal functions. To improve the translational potential of our approach, we tested intravitreal delivery of this system by means of adeno-associated viruses (AAVs). To this purpose, the employment of the AAV9-PHP.B resulted the most effective in disrupting the P23H Rho mutant. Finally, this approach was translated successfully in human cells engineered with the homozygous P23H RHO gene mutation. Overall, this is a significant proof-of-concept that gene allele specific targeting by CRISPR/Cas9 technology is specific and efficient and represents an unprecedented tool for treating RP and more broadly dominant genetic human disorders affecting the eye, as well as other tissues.


Assuntos
Marcação de Genes/métodos , Vetores Genéticos , Retina/fisiologia , Degeneração Retiniana/terapia , Rodopsina/genética , Alelos , Animais , Sistemas CRISPR-Cas , Eletroporação/métodos , Fibroblastos , Terapia Genética/métodos , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Mutação , RNA Guia de Cinetoplastídeos , Retina/patologia , Degeneração Retiniana/genética
3.
Mol Ther ; 25(12): 2727-2742, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-28882452

RESUMO

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.


Assuntos
Dependovirus/genética , Expressão Gênica , Vetores Genéticos/genética , beta-Glucosidase/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Eletroencefalografia , Ativação Enzimática , Ordem dos Genes , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Transdução Genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Sci Rep ; 6: 37540, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27857203

RESUMO

The CRISPR/Cas9 system is a rapid and customizable tool for gene editing in mammalian cells. In particular, this approach has widely opened new opportunities for genetic studies in neurological disease. Human neurons can be differentiated in vitro from hPSC (human Pluripotent Stem Cells), hNPCs (human Neural Precursor Cells) or even directly reprogrammed from fibroblasts. Here, we described a new platform which enables, rapid and efficient CRISPR/Cas9-mediated genome targeting simultaneously with three different paradigms for in vitro generation of neurons. This system was employed to inactivate two genes associated with neurological disorder (TSC2 and KCNQ2) and achieved up to 85% efficiency of gene targeting in the differentiated cells. In particular, we devised a protocol that, combining the expression of the CRISPR components with neurogenic factors, generated functional human neurons highly enriched for the desired genome modification in only 5 weeks. This new approach is easy, fast and that does not require the generation of stable isogenic clones, practice that is time consuming and for some genes not feasible.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/classificação , Sistemas CRISPR-Cas/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Inativação Gênica , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo
5.
EMBO Mol Med ; 8(10): 1197-1211, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27516453

RESUMO

Pantothenate kinase-associated neurodegeneration (PKAN) is an early onset and severely disabling neurodegenerative disease for which no therapy is available. PKAN is caused by mutations in PANK2, which encodes for the mitochondrial enzyme pantothenate kinase 2. Its function is to catalyze the first limiting step of Coenzyme A (CoA) biosynthesis. We generated induced pluripotent stem cells from PKAN patients and showed that their derived neurons exhibited premature death, increased ROS production, mitochondrial dysfunctions-including impairment of mitochondrial iron-dependent biosynthesis-and major membrane excitability defects. CoA supplementation prevented neuronal death and ROS formation by restoring mitochondrial and neuronal functionality. Our findings provide direct evidence that PANK2 malfunctioning is responsible for abnormal phenotypes in human neuronal cells and indicate CoA treatment as a possible therapeutic intervention.


Assuntos
Coenzima A/metabolismo , Neurônios/patologia , Neurodegeneração Associada a Pantotenato-Quinase/fisiopatologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Morte Celular , Células Cultivadas , Humanos , Mitocôndrias/patologia , Células-Tronco Pluripotentes/fisiologia , Espécies Reativas de Oxigênio/metabolismo
6.
Cell Stem Cell ; 17(6): 719-734, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26526726

RESUMO

Transplantation of GABAergic interneurons (INs) can provide long-term functional benefits in animal models of epilepsy and other neurological disorders. Whereas GABAergic INs can be differentiated from embryonic stem cells, alternative sources of GABAergic INs may be more tractable for disease modeling and transplantation. We identified five factors (Foxg1, Sox2, Ascl1, Dlx5, and Lhx6) that convert mouse fibroblasts into induced GABAergic INs (iGABA-INs) possessing molecular signatures of telencephalic INs. Factor overexpression activates transcriptional networks required for GABAergic fate specification. iGABA-INs display progressively maturing firing patterns comparable to cortical INs, form functional synapses, and release GABA. Importantly, iGABA-INs survive and mature upon being grafted into mouse hippocampus. Optogenetic stimulation demonstrated functional integration of grafted iGABA-INs into host circuitry, triggering inhibition of host granule neuron activity. These five factors also converted human cells into functional GABAergic INs. These properties suggest that iGABA-INs have potential for disease modeling and cell-based therapeutic approaches to neurological disorders.


Assuntos
Reprogramação Celular , Fibroblastos/citologia , Interneurônios/citologia , Prosencéfalo/citologia , Ácido gama-Aminobutírico/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Hipocampo/citologia , Humanos , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Fatores de Transcrição SOXB1/metabolismo , Sinapses/metabolismo , Telencéfalo/citologia , Transcrição Gênica
7.
Front Neurosci ; 8: 183, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25104921

RESUMO

New advances in directing the neuronal differentiation of human embryonic and induced pluripotent stem cells (hPSCs, abbreviation intended to convey both categories of pluripotent stem cells) have promoted the development of culture systems capable of modeling early neurogenesis and neural specification at some of their critical milestones. The hPSC-derived neural rosette can be considered the in vitro counterpart of the developing neural tube, since both structures share a virtually equivalent architecture and related functional properties. Epigenetic stimulation methods can modulate the identity of the rosette neural progenitors in order to generate authentic neuronal subtypes, as well as a full spectrum of neural crest derivatives. The intrinsic capacity of induced pluripotent cell-derived neural tissue to self-organize has become fully apparent with the emergence of innovative in vitro systems that are able to shape the neuronal differentiation of hPSCs into organized tissues that develop in three dimensions. However, significant hurdles remain that must be completely solved in order to facilitate the use of hPSCs in modeling (e.g., late-onset disorders) or in building therapeutic strategies for cell replacement. In this direction, new procedures have been established to promote the maturation and functionality of hPSC-derived neurons. Meanwhile, new methods to accelerate the aging of in vitro differentiating cells are still in development. hPSC-based technology has matured enough to offer a significant and reliable model system for early and late neurogenesis that could be extremely informative for the study of the physiological and pathological events that occur during this process. Thus, full exploitation of this cellular system can provide a better understanding of the physiological events that shape human brain structures, as well as a solid platform to investigate the pathological mechanisms at the root of human diseases.

8.
Stem Cells ; 29(2): 344-56, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21732491

RESUMO

There is growing evidence that Müller glia cells (MGCs) might act as regenerative elements in injured retinas of fishes and amniotes. However, their differentiation potential in humans is yet unknown. We isolated Müller glia from adult human retinas and propagated them in vitro revealing for the first time their ability to differentiate into rod photoreceptors. These results were also confirmed with mice retinas. Here, we describe conditions by which human MGCs adopt a rod photoreceptor commitment with a surprising efficiency as high as 54%. Functional characterization of Müller glia-derived photoreceptors by patch-clamp recordings revealed that their electrical properties are comparable to those of adult rods. Interestingly, our procedure allowed efficient derivation of MGC cultures starting from both injured and degenerating and postmortem human retinas. Human transplanted Müller glia-derived photoreceptors integrate and survive within immunodeficient mouse retinas. These data provide evidence that Müller glia retains an unpredicted plasticity and multipotent potential into adulthood, and it is therefore a promising source of novel therapeutic applications in retinal repair.


Assuntos
Neuroglia/citologia , Neuroglia/fisiologia , Regeneração , Retina , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Adulto , Animais , Diferenciação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Eletrofisiologia , Perfilação da Expressão Gênica , Gliose , Humanos , Camundongos , Técnicas de Patch-Clamp , Retina/citologia , Retina/lesões , Retina/transplante
9.
Exp Cell Res ; 316(7): 1148-58, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20171210

RESUMO

In this study we demonstrated that neural rosettes derived from human ES cells can give rise either to neural crest precursors, following expansion in presence of bFGF and EGF, or to dopaminergic precursors after exposure to ventralizing factors Shh and FGF8. Both regionalised precursors are capable of extensive proliferation and differentiation towards the corresponding terminally differentiated cell types. In particular, peripheral neurons, cartilage, bone, smooth muscle cells and also pigmented cells were obtained from neural crest precursors while tyrosine hydroxylase and Nurr1 positive dopaminergic neurons were derived from FGF8 and Shh primed rosette cells. Gene expression and immunocytochemistry analyses confirmed the expression of dorsal and neural crest genes such as Sox10, Slug, p75, FoxD3, Pax7 in neural precursors from bFGF-EGF exposed rosettes. By contrast, priming of rosettes with FGF8 and Shh induced the expression of dopaminergic markers Engrailed1, Pax2, Pitx3, floor plate marker FoxA2 and radial glia markers Blbp and Glast, the latter in agreement with the origin of dopaminergic precursors from floor plate radial glia. Moreover, in vivo transplant of proliferating Shh/FGF8 primed precursors in parkinsonian rats demonstrated engraftment and terminal dopaminergic differentiation. In conclusion, we demonstrated the derivation of long-term self-renewing precursors of selected regional identity as potential cell reservoirs for cell therapy applications, such as CNS degenerative diseases, or for the development of toxicological tests.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/farmacologia , Proteínas Hedgehog/farmacologia , Placa Neural/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Padronização Corporal/efeitos dos fármacos , Técnicas de Cultura de Células , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Sistema Nervoso Central/embriologia , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Placa Neural/embriologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurônios/transplante , Ratos , Ratos Sprague-Dawley , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/fisiologia , Esferoides Celulares/transplante , Células-Tronco/fisiologia , Fatores de Tempo , Transplante Heterólogo
10.
Stem Cells ; 24(11): 2514-21, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16931774

RESUMO

Embryonic stem cells differentiate into neuroectodermal cells under specific culture conditions. In primates, these cells are organized into rosettes expressing Pax6 and Sox1 and are responsive to inductive signals such as Sonic hedgehog (Shh) and retinoic acid. However, direct derivation of organized neuroectoderm in vitro from preimplantation mammalian embryos has never been reported. Here, we show that bovine inner cell masses from nuclear transfer and fertilized embryos, grown on feeders in serum-free medium, form polarized rosette structures expressing nestin, Pax6, Pax7, Sox1, and Otx2 and exhibiting interkinetic nuclear migration activity and cell junction distribution as in the developing neural tube. After in vitro expansion, neural rosettes give rise to p75-positive neural crest precursor cell lines capable of long-term proliferation and differentiation in autonomic and sensory peripheral neurons, glial cells, melanocytes, smooth muscle cells, and chondrocytes, recapitulating in vitro the unique plasticity of the neural crest lineage. Challenging the rosette dorsal fate by early exposure to Shh induces the expression of ventral markers Isl1, Nkx2.2, and Nkx6.1 and differentiation of mature astrocytes and neurons of central nervous system ventral identity, demonstrating appropriate response to inductive signals. All together, these findings indicate that neural rosettes directly derived from cloned and fertilized bovine embryos represent an in vitro model of early neural specification and differentiation events. Moreover, this study provides a source of highly proliferative neural crest precursor cell lines of wide differentiation potential for cell therapy and tissue engineering applications.


Assuntos
Astrócitos , Massa Celular Interna do Blastocisto , Linhagem da Célula , Células-Tronco Embrionárias , Crista Neural/citologia , Neurônios , Animais , Astrócitos/metabolismo , Astrócitos/transplante , Massa Celular Interna do Blastocisto/metabolismo , Bovinos , Diferenciação Celular , Proliferação de Células , Forma Celular , Células Cultivadas , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Células Clonais , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteína Homeobox Nkx-2.2 , Fator de Transcrição MSX1/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Crista Neural/metabolismo , Tumores Neuroectodérmicos/patologia , Neurônios/metabolismo , Neurônios/transplante , Fator de Transcrição PAX7/metabolismo , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXE , Transdução de Sinais , Fatores de Transcrição da Família Snail , Transplante de Células-Tronco , Fatores de Transcrição/metabolismo
11.
Stem Cells ; 24(4): 825-34, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16339994

RESUMO

Reliable procedures to induce neural commitment of totipotent undifferentiated embryonic stem (ES) cells have provided new tools for investigating the molecular mechanisms underlying cell fate choices. We extensively characterized the developmental potential of ES-induced neural cells obtained using an adaptation of the multistep induction protocol. We provided evidence that ES-derived neural proliferating cells are endowed with stem cell properties such as extensive self-renewal capacity and single-cell multipotency. In differentiating conditions, cells matured exclusively into neurons, astrocytes, and oligodendrocytes. All these features have been previously described in only somatic neural stem cells (NSCs). Therefore, we consider it more appropriate to rename our cells ES-derived NSCs. These similarities between the two NSC populations induced us to carefully compare their proliferation ability and differentiation potential. Although they were very similar in overall behavior, we scored specific differences. For instance, ES-derived NSCs proliferated at higher rate and consistently generated a higher number of neurons compared with somatic NSCs. To further investigate their relationships, we carried out a molecular analysis comparing their transcriptional profiles during proliferation. We observed a large fraction of shared expressed transcripts, including genes previously described to be critical in defining somatic NSC traits. Among the genes differently expressed, candidate genes possibly responsible for divergences between the two cell types were selected and further investigated. In particular, we showed that an enhanced MAPK (mitogen-activated protein kinase) signaling is acting in ES-induced NSCs, probably triggered by insulin-like growth factor-II. This may contribute to the high proliferation rate exhibited by these cells in culture.


Assuntos
Células-Tronco Multipotentes/citologia , Neurônios/citologia , Células-Tronco Totipotentes/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/citologia , Perfilação da Expressão Gênica , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Células-Tronco Multipotentes/metabolismo , Neurônios/metabolismo , Fenótipo , Transdução de Sinais , Células-Tronco Totipotentes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...