Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Small ; 19(30): e2205871, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37058009

RESUMO

Mechanical stimulation modulates neural development and neuronal activity. In a previous study, magnetic "nano-pulling" is proposed as a tool to generate active forces. By loading neural cells with magnetic nanoparticles (MNPs), a precise force vector is remotely generated through static magnetic fields. In the present study, human neural stem cells (NSCs) are subjected to a standard differentiation protocol, in the presence or absence of nano-pulling. Under mechanical stimulation, an increase in the length of the neural processes which showed an enrichment in microtubules, endoplasmic reticulum, and mitochondria is found. A stimulation lasting up to 82 days induces a strong remodeling at the level of synapse density and a re-organization of the neuronal network, halving the time required for the maturation of neural precursors into neurons. The MNP-loaded NSCs are then transplanted into mouse spinal cord organotypic slices, demonstrating that nano-pulling stimulates the elongation of the NSC processes and modulates their orientation even in an ex vivo model. Thus, it is shown that active mechanical stimuli can guide the outgrowth of NSCs transplanted into the spinal cord tissue. The findings suggest that mechanical forces play an important role in neuronal maturation which could be applied in regenerative medicine.


Assuntos
Células-Tronco Neurais , Traumatismos da Medula Espinal , Camundongos , Animais , Humanos , Neurônios , Medula Espinal/fisiologia , Diferenciação Celular/fisiologia , Neurogênese , Células Cultivadas
2.
Cereb Cortex ; 29(5): 2115-2124, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29688344

RESUMO

DACH1 is the human homolog of the Drosophila dachshund gene, which is involved in the development of the eye, nervous system, and limbs in the fly. Here, we systematically investigate DACH1 expression patterns during human neurodevelopment, from 5 to 21 postconceptional weeks. By immunodetection analysis, we found that DACH1 is highly expressed in the proliferating neuroprogenitors of the developing cortical ventricular and subventricular regions, while it is absent in the more differentiated cortical plate. Single-cell global transcriptional analysis revealed that DACH1 is specifically enriched in neuroepithelial and ventricular radial glia cells of the developing human neocortex. Moreover, we describe a previously unreported DACH1 expression in the human striatum, in particular in the striatal medium spiny neurons. This finding qualifies DACH1 as a new striatal projection neuron marker, together with PPP1R1B, BCL11B, and EBF1. We finally compared DACH1 expression profile in human and mouse forebrain, where we observed spatio-temporal similarities in its expression pattern thus providing a precise developmental description of DACH1 in the 2 mammalian species.


Assuntos
Corpo Estriado/embriologia , Corpo Estriado/metabolismo , Proteínas do Olho/metabolismo , Neocórtex/embriologia , Neocórtex/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Feto Abortado/embriologia , Feto Abortado/metabolismo , Células Ependimogliais/metabolismo , Idade Gestacional , Humanos , Ventrículos Laterais/embriologia , Ventrículos Laterais/metabolismo , Células-Tronco Neurais/metabolismo , Células Neuroepiteliais/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Especificidade da Espécie
3.
Dev Biol ; 411(1): 25-37, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26806704

RESUMO

HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells.


Assuntos
Diferenciação Celular/genética , Transição Epitelial-Mesenquimal/genética , Proteína HMGA2/fisiologia , Crista Neural/embriologia , Proteínas de Xenopus/fisiologia , Xenopus laevis/embriologia , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Proteína HMGA2/genética , Fator de Transcrição MSX1/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Morfolinos/genética , Crista Neural/citologia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Xenopus/genética
4.
Development ; 140(2): 301-12, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23250204

RESUMO

Medium-sized spiny neurons (MSNs) are the only neostriatum projection neurons, and their degeneration underlies some of the clinical features of Huntington's disease. Using knowledge of human developmental biology and exposure to key neurodevelopmental molecules, human pluripotent stem (hPS) cells were induced to differentiate into MSNs. In a feeder-free adherent culture, ventral telencephalic specification is induced by BMP/TGFß inhibition and subsequent SHH/DKK1 treatment. The emerging FOXG1(+)/GSX2(+) telencephalic progenitors are then terminally differentiated, resulting in the systematic line-independent generation of FOXP1(+)/FOXP2(+)/CTIP2(+)/calbindin(+)/DARPP-32(+) MSNs. Similar to mature MSNs, these neurons carry dopamine and A2a receptors, elicit a typical firing pattern and show inhibitory postsynaptic currents, as well as dopamine neuromodulation and synaptic integration ability in vivo. When transplanted into the striatum of quinolinic acid-lesioned rats, hPS-derived neurons survive and differentiate into DARPP-32(+) neurons, leading to a restoration of apomorphine-induced rotation behavior. In summary, hPS cells can be efficiently driven to acquire a functional striatal fate using an ontogeny-recapitulating stepwise method that represents a platform for in vitro human developmental neurobiology studies and drug screening approaches.


Assuntos
Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Sobrevivência Celular , Transplante de Células , Células-Tronco Embrionárias/citologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Neurônios GABAérgicos/metabolismo , Humanos , Doença de Huntington/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Técnicas de Patch-Clamp , Ácido Quinolínico/farmacologia , RNA/metabolismo , Ratos , Células-Tronco/citologia , Fatores de Tempo
5.
J Vis Exp ; (206)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38682940

RESUMO

Resolutive cures for spinal cord injuries (SCIs) are still lacking, due to the complex pathophysiology. One of the most promising regenerative approaches is based on stem cell transplantation to replace lost tissue and promote functional recovery. This approach should be further explored better in vitro and ex vivo for safety and efficacy before proceeding with more expensive and time-consuming animal testing. In this work, we show the establishment of a long-term platform based on mouse spinal cord (SC) organotypic slices transplanted with human neural stem cells to test cellular replacement therapies for SCIs. Standard SC organotypic cultures are maintained for around 2 or 3 weeks in vitro. Here, we describe an optimized protocol for long-term maintenance (≥30 days) for up to 90 days. The medium used for long-term culturing of SC slices was also optimized for transplanting neural stem cells into the organotypic model. Human SC-derived neuroepithelial stem (h-SC-NES) cells carrying a green fluorescent protein (GFP) reporter were transplanted into mouse SC slices. Thirty days after the transplant, cells still show GFP expression and a low apoptotic rate, suggesting that the optimized environment sustained their survival and integration inside the tissue. This protocol represents a robust reference for efficiently testing cell replacement therapies in the SC tissue. This platform will allow researchers to perform an ex vivo pre-screening of different cell transplantation therapies, helping them to choose the most appropriate strategy before proceeding with in vivo experiments.


Assuntos
Células-Tronco Neurais , Traumatismos da Medula Espinal , Medula Espinal , Animais , Camundongos , Traumatismos da Medula Espinal/terapia , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Medula Espinal/citologia , Técnicas de Cultura de Órgãos/métodos , Transplante de Células-Tronco/métodos
6.
Antiviral Res ; 223: 105816, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38286212

RESUMO

Human cytomegalovirus (HCMV) is the viral leading cause of congenital defects in newborns worldwide. Many aspects of congenital CMV (cCMV) infection, which currently lacks a specific treatment, as well as the main determinants of neuropathogenesis in the developing brain during HCMV infection are unclear. In this study, we modeled HCMV infection at different stages of neural development. Moreover, we evaluated the effects of both approved and investigational anti-HCMV drugs on viral replication and gene expression in two different neural progenitor cell lines, i.e., human embryonic stem cells-derived neural stem cells (NSCs) and fetus-derived neuroepithelial stem (NES) cells. Ganciclovir, letermovir, nitazoxanide, and the ozonide OZ418 reduced viral DNA synthesis and the production of infectious virus in both lines of neural progenitors. HCMV infection dysregulated the expression of genes that either are markers of neural progenitors, such as SOX2, NESTIN, PAX-6, or play a role in neurogenesis, such as Doublecortin. Treatment with antiviral drugs had different effects on HCMV-induced dysregulation of the genes under investigation. This study contributes to the understanding of the molecular mechanisms of cCMV neuropathogenesis and paves the way for further consideration of anti-HCMV drugs as candidate therapeutic agents for the amelioration of cCMV-associated neurological manifestations.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Recém-Nascido , Humanos , Infecções por Citomegalovirus/tratamento farmacológico , Encéfalo , Drogas em Investigação , Células-Tronco , Antivirais/farmacologia
7.
Front Neuroanat ; 17: 1130797, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36935652

RESUMO

The human brain is the most complex structure generated during development. Unveiling the ontogenesis and the intrinsic organization of specific neural networks may represent a key to understanding the physio-pathological aspects of different brain areas. The cortico-thalamic and thalamo-cortical (CT-TC) circuits process and modulate essential tasks such as wakefulness, sleep and memory, and their alterations may result in neurodevelopmental and psychiatric disorders. These pathologies are reported to affect specific neural populations but may also broadly alter physiological connections and thus dysregulate brain network generation, communication, and function. More specifically, the CT-TC system is reported to be severely affected in disorders impacting superior brain functions, such as schizophrenia (SCZ), bipolar disorder, autism spectrum disorders or epilepsy. In this review, the focus will be on CT development, and the models exploited to uncover and comprehend its molecular and cellular mechanisms. In parallel to animal models, still fundamental to unveil human neural network establishment, advanced in vitro platforms, such as brain organoids derived from human pluripotent stem cells, will be discussed. Indeed, organoids and assembloids represent unique tools to study and accelerate fundamental research in CT development and its dysfunctions. We will then discuss recent cutting-edge contributions, including in silico approaches, concerning ontogenesis, specification, and function of the CT-TC circuitry that generates connectivity maps in physiological and pathological conditions.

8.
Elife ; 122023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37272619

RESUMO

WDR62 is a spindle pole-associated scaffold protein with pleiotropic functions. Recessive mutations in WDR62 cause structural brain abnormalities and account for the second most common cause of autosomal recessive primary microcephaly (MCPH), indicating WDR62 as a critical hub for human brain development. Here, we investigated WDR62 function in corticogenesis through the analysis of a C-terminal truncating mutation (D955AfsX112). Using induced Pluripotent Stem Cells (iPSCs) obtained from a patient and his unaffected parent, as well as isogenic corrected lines, we generated 2D and 3D models of human neurodevelopment, including neuroepithelial stem cells, cerebro-cortical progenitors, terminally differentiated neurons, and cerebral organoids. We report that WDR62 localizes to the Golgi apparatus during interphase in cultured cells and human fetal brain tissue, and translocates to the mitotic spindle poles in a microtubule-dependent manner. Moreover, we demonstrate that WDR62 dysfunction impairs mitotic progression and results in alterations of the neurogenic trajectories of iPSC neuroderivatives. In summary, impairment of WDR62 localization and function results in severe neurodevelopmental abnormalities, thus delineating new mechanisms in the etiology of MCPH.


Assuntos
Proteínas de Ciclo Celular , Complexo de Golgi , Microcefalia , Proteínas do Tecido Nervoso , Polos do Fuso , Humanos , Microcefalia/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ciclo Celular/metabolismo , Masculino , Células-Tronco Pluripotentes Induzidas , Mitose , Criança , Adolescente
9.
Antiviral Res ; 216: 105664, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37414288

RESUMO

Recent evidence suggests that lipids play a crucial role in viral infections beyond their traditional functions of supplying envelope and energy, and creating protected niches for viral replication. In the case of Zika virus (ZIKV), it alters host lipids by enhancing lipogenesis and suppressing ß-oxidation to generate viral factories at the endoplasmic reticulum (ER) interface. This discovery prompted us to hypothesize that interference with lipogenesis could serve as a dual antiviral and anti-inflammatory strategy to combat the replication of positive sense single-stranded RNA (ssRNA+) viruses. To test this hypothesis, we examined the impact of inhibiting N-Acylethanolamine acid amidase (NAAA) on ZIKV-infected human Neural Stem Cells. NAAA is responsible for the hydrolysis of palmitoylethanolamide (PEA) in lysosomes and endolysosomes. Inhibition of NAAA results in PEA accumulation, which activates peroxisome proliferator-activated receptor-α (PPAR-α), directing ß-oxidation and preventing inflammation. Our findings indicate that inhibiting NAAA through gene-editing or drugs moderately reduces ZIKV replication by approximately one log10 in Human Neural Stem Cells, while also releasing immature virions that have lost their infectivity. This inhibition impairs furin-mediated prM cleavage, ultimately blocking ZIKV maturation. In summary, our study highlights NAAA as a host target for ZIKV infection.


Assuntos
Infecção por Zika virus , Zika virus , Humanos , Amidoidrolases/antagonistas & inibidores , Amidoidrolases/metabolismo , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Infecção por Zika virus/tratamento farmacológico
10.
Neurobiol Dis ; 46(1): 30-40, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22227000

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an excessive expansion of a CAG trinucleotide repeat in the gene encoding the protein huntingtin, resulting in an elongated stretch of glutamines near the N-terminus of the protein. Here we report the derivation of a collection of 11 induced pluripotent stem (iPS) cell lines generated through somatic reprogramming of fibroblasts obtained from the R6/2 transgenic HD mouse line. We show that CAG expansion has no effect on reprogramming efficiency, cell proliferation rate, brain-derived neurotrophic factor level, or neurogenic potential. However, genes involved in the cholesterol biosynthesis pathway, which is altered in HD, are also affected in HD-iPS cell lines. Furthermore, we found a lysosomal gene upregulation and an increase in lysosome number in HD-iPS cell lines. These observations suggest that iPS cells from HD mice replicate some but not all of the molecular phenotypes typically observed in the disease; additionally, they do not manifest increased cell death propensity either under self-renewal or differentiated conditions. More studies will be necessary to transform a revolutionary technology into a powerful platform for drug screening approaches.


Assuntos
Diferenciação Celular/genética , Doença de Huntington/enzimologia , Células-Tronco Pluripotentes Induzidas/enzimologia , Lisossomos/enzimologia , Neurônios/enzimologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Proteína Huntingtina , Doença de Huntington/genética , Células-Tronco Pluripotentes Induzidas/citologia , Lisossomos/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/citologia , Vias Neurais/enzimologia , Neurônios/citologia , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Cultura Primária de Células
11.
Cell Mol Life Sci ; 68(10): 1769-83, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20981563

RESUMO

Neural stem (NS) cells are a self-renewing population of symmetrically dividing multipotent radial glia-like stem cells, characterized by homogeneous expansion in monolayer. Here we report that fetal NS cells isolated from different regions of the developing mouse nervous system behave in a similar manner with respect to self-renewal and neuropotency, but exhibit distinct positional identities. For example, NS cells from the neocortex maintain the expression of anterior transcription factors, including Otx2 and Foxg1, while Hoxb4 and Hoxb9 are uniquely found in spinal cord-derived NS cells. This molecular signature was stable for over 20 passages and was strictly linked to the developmental stage of the donor, because only NS cells derived from E14.5 cortex, and not those derived from E12.5 cortex, carried a consistent transcription factor profile. We also showed that traits of this positional code are maintained during neuronal differentiation, leading to the generation of electrophysiologically active neurons, even if they do not acquire a complete neurochemical identity.


Assuntos
Feto/citologia , Células-Tronco Neurais/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Neocórtex/citologia , Neocórtex/embriologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Técnicas de Patch-Clamp , Medula Espinal/citologia , Medula Espinal/embriologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Sci Rep ; 12(1): 2341, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149717

RESUMO

The growth of glioblastoma (GBM), one of the deadliest adult cancers, is fuelled by a subpopulation of stem/progenitor cells, which are thought to be the source of resistance and relapse after treatment. Re-engagement of a latent capacity of these cells to re-enter a trajectory resulting in cell differentiation is a potential new therapeutic approach for this devastating disease. ASCL1, a proneural transcription factor, plays a key role in normal brain development and is also expressed in a subset of GBM cells, but fails to engage a full differentiation programme in this context. Here, we investigated the barriers to ASCL1-driven differentiation in GBM stem cells. We see that ASCL1 is highly phosphorylated in GBM stem cells where its expression is compatible with cell proliferation. However, overexpression of a form of ASCL1 that cannot be phosphorylated on Serine-Proline sites drives GBM cells down a neuronal lineage and out of cell cycle more efficiently than its wild-type counterpart, an effect further enhanced by deletion of the inhibitor of differentiation ID2, indicating mechanisms to reverse the block to GBM cell differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Glioblastoma/metabolismo , Glioblastoma/fisiopatologia , Proteína 2 Inibidora de Diferenciação/genética , Células-Tronco Neoplásicas/metabolismo , Motivos de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias Encefálicas/genética , Ciclo Celular , Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Proteína 2 Inibidora de Diferenciação/metabolismo , Células-Tronco Neoplásicas/citologia , Fosforilação
14.
Stem Cell Reports ; 17(7): 1683-1698, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35714598

RESUMO

Congenital alterations in the levels of the transcription factor Forkhead box g1 (FOXG1) coding gene trigger "FOXG1 syndrome," a spectrum that recapitulates birth defects found in the "congenital Zika syndrome," such as microcephaly and other neurodevelopmental conditions. Here, we report that Zika virus (ZIKV) infection alters FOXG1 nuclear localization and causes its downregulation, thus impairing expression of genes involved in cell replication and apoptosis in several cell models, including human neural progenitor cells. Growth factors, such as EGF and FGF2, and Thr271 residue located in FOXG1 AKT domain, take part in the nuclear displacement and apoptosis protection, respectively. Finally, by progressive deletion of FOXG1 sequence, we identify the C-terminus and the residues 428-481 as critical domains. Collectively, our data suggest a causal mechanism by which ZIKV affects FOXG1, its target genes, cell cycle progression, and survival of human neural progenitors, thus contributing to microcephaly.


Assuntos
Microcefalia , Células-Tronco Neurais , Infecção por Zika virus , Zika virus , Regulação para Baixo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Microcefalia/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Zika virus/fisiologia , Infecção por Zika virus/genética
15.
Front Cell Neurosci ; 16: 858347, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35573835

RESUMO

As microtubule-organizing centers (MTOCs), centrosomes play a pivotal role in cell division, neurodevelopment and neuronal maturation. Among centrosomal proteins, centrin-2 (CETN2) also contributes to DNA repair mechanisms which are fundamental to prevent genomic instability during neural stem cell pool expansion. Nevertheless, the expression profile of CETN2 in human neural stem cells and their progeny is currently unknown. To address this question, we interrogated a platform of human neuroepithelial stem (NES) cells derived from post mortem developing brain or established from pluripotent cells and demonstrated that while CETN2 retains its centrosomal location in proliferating NES cells, its expression pattern changes upon differentiation. In particular, we found that CETN2 is selectively expressed in mature astrocytes with a broad cytoplasmic distribution. We then extended our findings on human autoptic nervous tissue samples. We investigated CETN2 distribution in diverse anatomical areas along the rostro-caudal neuraxis and pointed out a peculiar topography of CETN2-labeled astrocytes in humans which was not appreciable in murine tissues, where CETN2 was mostly confined to ependymal cells. As a prototypical condition with glial overproliferation, we also explored CETN2 expression in glioblastoma multiforme (GBM), reporting a focal concentration of CETN2 in neoplastic astrocytes. This study expands CETN2 localization beyond centrosomes and reveals a unique expression pattern that makes it eligible as a novel astrocytic molecular marker, thus opening new roads to glial biology and human neural conditions.

16.
Mol Cell Neurosci ; 43(3): 287-95, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20026276

RESUMO

Neural stem (NS) cells are a homogenous population of stem cells that expands in monolayer under serum-free conditions while remaining highly neuropotent. Here, we generated NS cells from induced pluripotent stem (iPS) cells that were previously derived from mouse fibroblasts (NS-(f)iPS). We showed that NS-(f)iPS cells exhibit long-term expansion and express markers of neurogenic radial glia. Analyses of the regional markers expressed in NS-(f)iPS cells suggested a ventral-rhombencephalic identity. Upon exposure to differentiation protocols, NS-(f)iPS cells produce neurons, astrocytes, and oligodendrocytes with an efficiency similar to ES-derived NS cells. NS-(f)iPS cells represent a new tool for studying neural cell fate determination and terminal differentiation, providing an interesting resource for experimental transplantation. Comparative studies between NS cells derived from iPS cells, reprogrammed from different somatic sources, and from authentic ES cells are necessary to identify critical elements for multipotency acquisition.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Neurônios/fisiologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Neurônios/citologia , Oligodendroglia/citologia , Oligodendroglia/fisiologia
17.
Prog Mol Biol Transl Sci ; 182: 403-438, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34175049

RESUMO

The recent advent of genome editing techniques and their rapid improvement paved the way in establishing innovative human neurological disease models and in developing new therapeutic opportunities. Human pluripotent (both induced or naive) stem cells and neural stem cells represent versatile tools to be applied to multiple research needs and, together with genomic snip and fix tools, have recently made possible the creation of unique platforms to directly investigate several human neural affections. In this chapter, we will discuss genome engineering tools, and their recent improvements, applied to the stem cell field, focusing on how these two technologies may be pivotal instruments to deeply unravel molecular mechanisms underlying development and function, as well as disorders, of the human brain. We will review how these frontier technologies may be exploited to investigate or treat severe neurodevelopmental disorders, such as microcephaly, autism spectrum disorder, schizophrenia, as well as neurodegenerative conditions, including Parkinson's disease, Huntington's disease, Alzheimer's disease, and spinal muscular atrophy.


Assuntos
Transtorno do Espectro Autista , Células-Tronco Pluripotentes Induzidas , Células-Tronco Neurais , Doenças Neurodegenerativas , Edição de Genes , Humanos
18.
Front Cell Dev Biol ; 9: 637565, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33718375

RESUMO

Snap29 is a conserved regulator of membrane fusion essential to complete autophagy and to support other cellular processes, including cell division. In humans, inactivating SNAP29 mutations causes CEDNIK syndrome, a rare multi-systemic disorder characterized by congenital neuro-cutaneous alterations. The fibroblasts of CEDNIK patients show alterations of the Golgi apparatus (GA). However, whether and how Snap29 acts at the GA is unclear. Here we investigate SNAP29 function at the GA and endoplasmic reticulum (ER). As part of the elongated structures in proximity to these membrane compartments, a pool of SNAP29 forms a complex with Syntaxin18, or with Syntaxin5, which we find is required to engage SEC22B-loaded vesicles. Consistent with this, in HeLa cells, in neuroepithelial stem cells, and in vivo, decreased SNAP29 activity alters GA architecture and reduces ER to GA trafficking. Our data reveal a new regulatory function of Snap29 in promoting secretory trafficking.

19.
Cells ; 9(8)2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32806773

RESUMO

Building and functioning of the human brain requires the precise orchestration and execution of myriad molecular and cellular processes, across a multitude of cell types and over an extended period of time. Dysregulation of these processes affects structure and function of the brain and can lead to neurodevelopmental, neurological, or psychiatric disorders. Multiple environmental stimuli affect neural stem cells (NSCs) at several levels, thus impairing the normal human neurodevelopmental program. In this review article, we will delineate the main mechanisms of infection adopted by several neurotropic pathogens, and the selective NSC vulnerability. In particular, TORCH agents, i.e., Toxoplasma gondii, others (including Zika virus and Coxsackie virus), Rubella virus, Cytomegalovirus, and Herpes simplex virus, will be considered for their devastating effects on NSC self-renewal with the consequent neural progenitor depletion, the cellular substrate of microcephaly. Moreover, new evidence suggests that some of these agents may also affect the NSC progeny, producing long-term effects in the neuronal lineage. This is evident in the paradigmatic example of the neurodegeneration occurring in Alzheimer's disease.


Assuntos
Doença de Alzheimer/parasitologia , Doença de Alzheimer/virologia , Microcefalia/parasitologia , Microcefalia/virologia , Células-Tronco Neurais/parasitologia , Células-Tronco Neurais/virologia , Transtornos do Neurodesenvolvimento/parasitologia , Transtornos do Neurodesenvolvimento/virologia , Animais , Infecções por Vírus de DNA/complicações , Infecções por Vírus de DNA/virologia , Vírus de DNA/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Infecções por Vírus de RNA/complicações , Infecções por Vírus de RNA/virologia , Vírus de RNA/patogenicidade , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia , Virulência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa