Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 19(2): e1010606, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36745687

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder caused by progressive loss of motor neurons and there is currently no effective therapy. Cytoplasmic mislocalization and aggregation of TAR DNA-binding protein 43 kDa (TDP-43) within the CNS is a pathological hallmark in sporadic ALS and prion-like propagation of pathogenic TDP-43 is thought to be implicated in disease progression. However, cell-to-cell transmission of pathogenic TDP-43 in the human CNS has not been confirmed experimentally. Here we used induced pluripotent stem cells (iPSCs)-derived cerebral organoids as recipient CNS tissue model that are anatomically relevant human brain. We injected postmortem spinal cord protein extracts individually from three non-ALS or five sporadic ALS patients containing pathogenic TDP-43 into the cerebral organoids to validate the templated propagation and spreading of TDP-43 pathology in human CNS tissue. We first demonstrated that the administration of spinal cord extracts from an ALS patient induced the formation of TDP-43 pathology that progressively spread in a time-dependent manner in cerebral organoids, suggesting that pathogenic TDP-43 from ALS functioned as seeds and propagated cell-to-cell to form de novo TDP-43 pathology. We also reported that the administration of ALS patient-derived protein extracts caused astrocyte proliferation to form astrogliosis in cerebral organoids, reproducing the pathological feature seen in ALS. Moreover, we showed pathogenic TDP-43 induced cellular apoptosis and that TDP-43 pathology correlated with genomic damage due to DNA double-strand breaks. Thus, our results provide evidence that patient-derived pathogenic TDP-43 can mimic the prion-like propagation of TDP-43 pathology in human CNS tissue. Our findings indicate that our assays with human cerebral organoids that replicate ALS pathophysiology have a promising strategy for creating readouts that could be used in future drug discovery efforts against ALS.


Assuntos
Esclerose Lateral Amiotrófica , Príons , Humanos , Esclerose Lateral Amiotrófica/patologia , Medula Espinal/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Príons/metabolismo , Organoides/metabolismo
2.
EMBO J ; 37(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29903919

RESUMO

Glioblastoma is the most common and aggressive brain tumor, with a subpopulation of stem-like cells thought to mediate its recurring behavior and therapeutic resistance. The epithelial-mesenchymal transition (EMT) inducing factor Zeb1 was linked to tumor initiation, invasion, and resistance to therapy in glioblastoma, but how Zeb1 functions at molecular level and what genes it regulates remain poorly understood. Contrary to the common view that EMT factors act as transcriptional repressors, here we show that genome-wide binding of Zeb1 associates with both activation and repression of gene expression in glioblastoma stem-like cells. Transcriptional repression requires direct DNA binding of Zeb1, while indirect recruitment to regulatory regions by the Wnt pathway effector Lef1 results in gene activation, independently of Wnt signaling. Amongst glioblastoma genes activated by Zeb1 are predicted mediators of tumor cell migration and invasion, including the guanine nucleotide exchange factor Prex1, whose elevated expression is predictive of shorter glioblastoma patient survival. Prex1 promotes invasiveness of glioblastoma cells in vivo highlighting the importance of Zeb1/Lef1 gene regulatory mechanisms in gliomagenesis.


Assuntos
Glioblastoma/genética , Glioblastoma/patologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Via de Sinalização Wnt/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal/genética , Glioblastoma/mortalidade , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Invasividade Neoplásica/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
3.
J Neuroinflammation ; 16(1): 148, 2019 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-31319868

RESUMO

BACKGROUND: Neuroinflammation is the response of the central nervous system to events that interfere with tissue homeostasis and represents a common denominator in virtually all neurological diseases. Activation of microglia, the principal immune effector cells of the brain, contributes to neuronal injury by release of neurotoxic products. Toll-like receptor 4 (TLR4), expressed on the surface of microglia, plays an important role in mediating lipopolysaccharide (LPS)-induced microglia activation and inflammatory responses. We have previously shown that curcumin and some of its analogues harboring an α,ß-unsaturated 1,3-diketone moiety, able to coordinate the magnesium ion, can interfere with LPS-mediated TLR4-myeloid differentiation protein-2 (MD-2) signaling. Fluoroquinolone (FQ) antibiotics are compounds that contain a keto-carbonyl group that binds divalent ions, including magnesium. In addition to their antimicrobial activity, FQs are endowed with immunomodulatory properties, but the mechanism underlying their anti-inflammatory activity remains to be defined. The aim of the current study was to elucidate the molecular mechanism of these compounds in the TLR4/NF-κB inflammatory signaling pathway. METHODS: The putative binding mode of five FQs [ciprofloxacin (CPFX), levofloxacin (LVFX), moxifloxacin, ofloxacin, and delafloxacin] to TLR4-MD-2 was determined using molecular docking simulations. The effect of CPFX and LVFX on LPS-induced release of IL-1ß and TNF-α and NF-κB activation was investigated in primary microglia by ELISA and fluorescence staining. The interaction of CPFX and LVFX with TLR4-MD-2 complex was assessed by immunoprecipitation followed by Western blotting using Ba/F3 cells. RESULTS: CPFX and LVFX bound to the hydrophobic region of the MD-2 pocket and inhibited LPS-induced secretion of pro-inflammatory cytokines and activation of NF-κB in primary microglia. Furthermore, these FQs diminished the binding of LPS to TLR4-MD-2 complex and decreased the resulting TLR4-MD-2 dimerization in Ba/F3 cells. CONCLUSIONS: These results provide new insight into the mechanism of the anti-inflammatory activity of CPFX and LVFX, which involves, at least in part, the activation of TLR4/NF-κB signaling pathway. Our findings might facilitate the development of new molecules directed at the TLR4-MD-2 complex, a potential key target for controlling neuroinflammation.


Assuntos
Ciprofloxacina/farmacologia , Inflamação/imunologia , Levofloxacino/farmacologia , Microglia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Humanos , Inflamação/metabolismo , Camundongos , Microglia/imunologia , NF-kappa B/efeitos dos fármacos , NF-kappa B/imunologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/efeitos dos fármacos , Receptor 4 Toll-Like/imunologia
4.
Glia ; 66(12): 2659-2672, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30338559

RESUMO

Nuclear factor-kappaB (NF-κB) is activated in neural progenitor cells in the developing murine cerebral cortex during the neurogenic phase, when it acts to prevent premature neuronal differentiation. Here we show that NF-κB activation continues in mouse neocortical neural progenitor cells during the neurogenic-to-gliogenic switch. Blockade of endogenous NF-κB activity during neocortical gliogenesis leads to the formation of supernumerary committed gliogenic progenitors and premature glial cell differentiation. Conversely, forced NF-κB activation during the neocortical neurogenic-to-gliogenic transition causes delayed gliogenic commitment and decreased astroglial gene expression. NF-κB activation continues in neocortical gliogenic progenitors following commitment and is important to inhibit the differentiation of oligodendrocyte precursor cells and to maintain persistent expression of glial fibrillary acidic protein in maturing astrocytes. These results reveal a number of previously uncharacterized roles for NF-κB during different phases of neocortical gliogenesis and identify NF-κB as an inhibitor of early oligodendrocyte development in the cerebral cortex.


Assuntos
Córtex Cerebral , Regulação da Expressão Gênica no Desenvolvimento/genética , NF-kappa B/metabolismo , Neurogênese/genética , Neuroglia/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/embriologia , Ventrículos Cerebrais/crescimento & desenvolvimento , Fator Neurotrófico Ciliar/farmacologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/fisiologia
5.
Adv Exp Med Biol ; 962: 103-116, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28299654

RESUMO

Runt-related (Runx) transcription factors play essential roles during development and adult tissue homeostasis and are responsible for several human diseases. They regulate a variety of biological mechanisms in numerous cell lineages. Recent years have seen significant progress in our understanding of the functions performed by Runx proteins in the developing and postnatal mammalian nervous system. In both central and peripheral nervous systems, Runx1 and Runx3 display remarkably specific expression in mostly non-overlapping groups of postmitotic neurons. In the central nervous system, Runx1 is involved in the development of selected motor neurons controlling neural circuits mediating vital functions such as chewing, swallowing, breathing, and locomotion. In the peripheral nervous system, Runx1 and Runx3 play essential roles during the development of sensory neurons involved in circuits mediating pain, itch, thermal sensation and sense of relative position. Runx1 and Runx3 orchestrate complex gene expression programs controlling neuronal subtype specification and axonal connectivity. Runx1 is also important in the olfactory system, where it regulates the progenitor-to-neuron transition in undifferentiated neural progenitor cells in the olfactory epithelium as well as the proliferation and developmental maturation of specific glial cells termed olfactory ensheathing cells. Moreover, upregulated Runx expression is associated with brain injury and disease. Increasing knowledge of the functions of Runx proteins in the developing and postnatal nervous system is therefore expected to improve our understanding of nervous system development, homeostasis and disease.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/metabolismo , Organogênese/fisiologia , Animais , Linhagem da Célula/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos
6.
Stem Cells ; 32(6): 1591-601, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24307508

RESUMO

Although inhibition of p16(INK4a) expression is critical to preserve the proliferative capacity of stem cells, the molecular mechanisms responsible for silencing p16(INK4a) expression remain poorly characterized. Here, we show that the histone acetyltransferase (HAT) monocytic leukemia zinc finger protein (MOZ) controls the proliferation of both hematopoietic and neural stem cells by modulating the transcriptional repression of p16(INK4a) . In the absence of the HAT activity of MOZ, expression of p16(INK4a) is upregulated in progenitor and stem cells, inducing an early entrance into replicative senescence. Genetic deletion of p16(INK4a) reverses the proliferative defect in both Moz(HAT) (-) (/) (-) hematopoietic and neural progenitors. Our results suggest a critical requirement for MOZ HAT activity to silence p16(INK4a) expression and to protect stem cells from early entrance into replicative senescence.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células-Tronco Hematopoéticas/citologia , Histona Acetiltransferases/metabolismo , Células-Tronco Neurais/citologia , Animais , Antígenos CD34/metabolismo , Ciclo Celular , Proliferação de Células , Separação Celular , Senescência Celular , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Deleção de Genes , Inativação Gênica , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Telencéfalo/citologia , Regulação para Cima/genética
7.
BMC Cancer ; 14: 139, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24575771

RESUMO

BACKGROUND: Embryonal Rhabdomyosarcoma (RMS) is a pediatric soft-tissue sarcoma derived from myogenic precursors that is characterized by a good prognosis in patients with localized disease. Conversely, metastatic tumors often relapse, leading to a dismal outcome. The histone methyltransferase EZH2 epigenetically suppresses skeletal muscle differentiation by repressing the transcription of myogenic genes. Moreover, de-regulated EZH2 expression has been extensively implied in human cancers. We have previously shown that EZH2 is aberrantly over-expressed in RMS primary tumors and cell lines. Moreover, it has been recently reported that EZH2 silencing in RD cells, a recurrence-derived embryonal RMS cell line, favors myofiber-like structures formation in a pro-differentiation context. Here we evaluate whether similar effects can be obtained also in the presence of growth factor-supplemented medium (GM), that mimics a pro-proliferative microenvironment, and by pharmacological targeting of EZH2 in RD cells and in RD tumor xenografts. METHODS: Embryonal RMS RD cells were cultured in GM and silenced for EZH2 or treated with either the S-adenosylhomocysteine hydrolase inhibitor 3-deazaneplanocin A (DZNep) that induces EZH2 degradation, or with a new class of catalytic EZH2 inhibitors, MC1948 and MC1945, which block the catalytic activity of EZH2. RD cell proliferation and myogenic differentiation were evaluated both in vitro and in vivo. RESULTS: Here we show that EZH2 protein was abnormally expressed in 19 out of 19 (100%) embryonal RMS primary tumors and cell lines compared to their normal counterparts. Genetic down-regulation of EZH2 by silencing in GM condition reduced RD cell proliferation up-regulating p21Cip1. It also resulted in myogenic-like differentiation testified by the up-regulation of myogenic markers Myogenin, MCK and MHC. These effects were reverted by enforced over-expression of a murine Ezh2, highlighting an EZH2-specific effect. Pharmacological inhibition of EZH2 using either DZNep or MC inhibitors phenocopied the genetic knockdown of EZH2 preventing cell proliferation and restoring myogenic differentiation both in vitro and in vivo. CONCLUSIONS: These results provide evidence that EZH2 function can be counteracted by pharmacological inhibition in embryonal RMS blocking proliferation even in a pro-proliferative context. They also suggest that this approach could be exploited as a differentiation therapy in adjuvant therapeutic intervention for embryonal RMS.


Assuntos
Antineoplásicos/uso terapêutico , Complexo Repressor Polycomb 2/antagonistas & inibidores , Rabdomiossarcoma Embrionário/tratamento farmacológico , Rabdomiossarcoma Embrionário/metabolismo , Adolescente , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Criança , Pré-Escolar , Modelos Animais de Doenças , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Complexo Repressor Polycomb 2/metabolismo , Rabdomiossarcoma Embrionário/patologia , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Commun Biol ; 7(1): 238, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418587

RESUMO

The fatal motor neuron (MN) disease Amyotrophic Lateral Sclerosis (ALS) is characterized by progressive MN degeneration. Phrenic MNs (phMNs) controlling the activity of the diaphragm are prone to degeneration in ALS, leading to death by respiratory failure. Understanding of the mechanisms of phMN degeneration in ALS is limited, mainly because human experimental models to study phMNs are lacking. Here we describe a method enabling the derivation of phrenic-like MNs from human iPSCs (hiPSC-phMNs) within 30 days. This protocol uses an optimized combination of small molecules followed by cell-sorting based on a cell-surface protein enriched in hiPSC-phMNs, and is highly reproducible using several hiPSC lines. We show further that hiPSC-phMNs harbouring ALS-associated amplification of the C9orf72 gene progressively lose their electrophysiological activity and undergo increased death compared to isogenic controls. These studies establish a previously unavailable protocol to generate human phMNs offering a disease-relevant system to study mechanisms of respiratory MN dysfunction.


Assuntos
Esclerose Lateral Amiotrófica , Células-Tronco Pluripotentes Induzidas , Transtornos Respiratórios , Humanos , Esclerose Lateral Amiotrófica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/fisiologia , Diafragma , Transtornos Respiratórios/metabolismo , Degeneração Neural
9.
J Neurosci ; 32(33): 11285-98, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22895712

RESUMO

Microglia are the immune cells of the nervous system, where they act as resident macrophages during inflammatory events underlying many neuropathological conditions. Microglia derive from primitive myeloid precursors that colonize the nervous system during embryonic development. In the postnatal brain, microglia are initially mitotic, rounded in shape (amoeboid), and phagocytically active. As brain development proceeds, they gradually undergo a transition to a surveillant nonphagocytic state characterized by a highly branched (ramified) morphology. This ramification process is almost recapitulated in reverse during the process of microglia activation in the adult brain, when surveillant microglia undergo a ramified-to-amoeboid morphological transformation and become phagocytic in response to injury or disease. Little is known about the mechanisms controlling amoeboid microglial cell proliferation, activation, and ramification during brain development, despite the critical role of these processes in the establishment of the adult microglia pool and their relevance to microglia activation in the adult brain. Here we show that the mouse transcription factor Runx1, a key regulator of myeloid cell proliferation and differentiation, is expressed in forebrain amoeboid microglia during the first two postnatal weeks. Runx1 expression is then downregulated in ramified microglia. Runx1 inhibits mouse amoeboid microglia proliferation and promotes progression to the ramified state. We show further that Runx1 expression is upregulated in microglia following nerve injury in the adult mouse nervous system. These findings provide insight into the regulation of postnatal microglia activation and maturation to the ramified state and have implications for microglia biology in the developing and injured brain.


Assuntos
Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Microglia/metabolismo , Prosencéfalo/citologia , Prosencéfalo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação/metabolismo , Bromodesoxiuridina/metabolismo , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Transformada , Células Cultivadas , Imunoprecipitação da Cromatina , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Óxido Nítrico Sintase Tipo II/metabolismo , Fosfatidiletanolaminas , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Medula Espinal/citologia
10.
J Biol Chem ; 287(18): 14749-59, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22354967

RESUMO

Transducin-like enhancer of split-1 (TLE1) plays a critical role in the regulation of neurogenesis by inhibiting the differentiation of neural progenitor cells into neurons. Although TLE1 is also expressed highly in the postnatal brain and through adulthood, its role in postmitotic neurons is not clear. Using cultures of cerebellar granule neurons, we show that expression of TLE1 is reduced in neurons primed to die. Reestablishment of elevated TLE1 levels by ectopic expression protects neurons from death, whereas suppression of TLE1 expression in otherwise healthy neurons induces cell death. These results show that TLE1 is necessary for the maintenance of neuronal survival. Experiments using pharmacological inhibitors as well as expression of point mutants indicate that phosphorylation of TLE1 by casein kinase-2 (CK2) at Ser-239 and Ser-253 is necessary for its survival-promoting activity. TLE1-mediated survival is also inhibited by pharmacological inhibition of PI3K-Akt signaling but not by inhibitors of Raf-MEK-ERK signaling or other molecules, including histone deacetylases, calcium calmodulin kinase, or CK1. The survival-promoting activity of TLE1 depends critically on interaction with FoxG1, another protein involved in the regulation of neurogenesis and shown previously to promote survival of postmitotic neurons. Likewise, the ability of FoxG1 to promote neuronal survival depends on TLE1. Taken together, our study demonstrates that TLE1 cooperates with FoxG1 to promote neuronal survival in a CK2- and PI3K-Akt-dependent manner.


Assuntos
Cerebelo/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Animais , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Cerebelo/citologia , Fatores de Transcrição Forkhead/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Proteínas Repressoras/genética , Quinases raf/genética , Quinases raf/metabolismo
11.
Exp Cell Res ; 317(4): 464-73, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21093432

RESUMO

Krüppel-like factor 7 (KLF7) belongs to the large family of KLF transcription factors, which comprises at least 17 members. Within this family, KLF7 is unique since its expression is strictly restricted within the nervous system during development. We have previously shown that KLF7 is required for neuronal morphogenesis and axon guidance in selected regions of the nervous system, including hippocampus, olfactory bulbs and cortex, as well as in neuronal cell cultures. In the present work, we have furthered our analysis of the role of KLF7 in central nervous system development. By gene expression analysis during brain embryogenesis, we found significant alterations in dopaminergic neurons in Klf7 null mice. In particular, the tyrosine hydroxylase (TH) and dopamine transporter (Dat) transcripts are strongly decreased in the olfactory bulbs and ventral midbrain at birth, compared to wild-type littermates. Interestingly, Klf7-mutant mice show a dramatic reduction of TH-positive neurons in the olfactory bulbs, but no change in GABAergic or midbrain dopaminergic neurons. These observations raise the possibility that a lack of a KLF family member affects dopaminergic neuron development.


Assuntos
Dopamina , Desenvolvimento Embrionário , Fatores de Transcrição Kruppel-Like/fisiologia , Neurônios/citologia , Bulbo Olfatório/crescimento & desenvolvimento , Animais , Sistema Nervoso Central/crescimento & desenvolvimento , Proteínas da Membrana Plasmática de Transporte de Dopamina/biossíntese , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Bulbo Olfatório/citologia , Fatores de Transcrição/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese , Tirosina 3-Mono-Oxigenase/genética
12.
ASN Neuro ; 14: 17590914221145105, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36524236

RESUMO

As the resident immune cells of the healthy nervous system, homeostatic microglia can rapidly become activated in response to injury/disease. Dysregulated microglia activation is a hallmark of nervous system disorders including neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. The elucidation of the biological and pathological roles of microglia has recently benefitted from the development of microglia-like cells using human induced pluripotent stem cell (iPSC)-based approaches. The success of iPSC-derived microglia preparations as a disease-relevant model system depends on their representation of the in vivo spatial and temporal heterogeneity of microglia under pathological conditions. Little is currently known about the potential of human iPSC-derived microglia generated using different methods for the study of neurodegenerative diseases. We compared the transcriptomes of human iPSC-derived microglia generated using two frequently used in vitro differentiation methods to determine whether separate strategies can generate microglia with distinct transcriptional signatures in vitro. We show that microglia derived using different differentiation methods display distinct maturation characteristics after equivalent times in culture. We also reveal that iPSC-derived microglia preparations generated using these two methods are composed of different subpopulations with transcriptomic signatures resembling those of in vivo regionally distinct microglia subtypes, specifically white-matter and gray-matter microglia. These findings highlight the need to better characterize the subtype composition of each microglia preparation prior to its use to model neurodegenerative diseases.


Assuntos
Esclerose Lateral Amiotrófica , Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Microglia/patologia , Diferenciação Celular , Esclerose Lateral Amiotrófica/patologia
13.
ASN Neuro ; 14: 17590914211073381, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35023784

RESUMO

Human induced pluripotent stem cells (hiPSCs) derived from healthy and diseased individuals can give rise to many cell types, facilitating the study of mechanisms of development, human disease modeling, and early drug target validation. In this context, experimental model systems based on hiPSC-derived motor neurons (MNs) have been used to study MN diseases such as spinal muscular atrophy and amyotrophic lateral sclerosis. Modeling MN disease using hiPSC-based approaches requires culture conditions that can recapitulate in a dish the events underlying differentiation, maturation, aging, and death of MNs. Current hiPSC-derived MN-based applications are often hampered by limitations in our ability to monitor MN morphology, survival, and other functional properties over a prolonged timeframe, underscoring the need for improved long-term culture conditions. Here we describe a cytocompatible dendritic polyglycerol amine (dPGA) substrate-based method for prolonged culture of hiPSC-derived MNs. We provide evidence that MNs cultured on dPGA-coated dishes are more amenable to long-term study of cell viability, molecular identity, and spontaneous network electrophysiological activity. The present study has the potential to improve hiPSC-based studies of human MN biology and disease.We describe the use of a new coating substrate providing improved conditions for long-term cultures of human iPSC-derived motor neurons, thus allowing evaluation of cell viability, molecular identity, spontaneous network electrophysiological activity, and single-cell RNA sequencing of mature motor neurons.


Assuntos
Células-Tronco Pluripotentes Induzidas , Aminas , Diferenciação Celular , Glicerol , Humanos , Neurônios Motores , Polímeros
14.
Cells ; 11(3)2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35159209

RESUMO

Astrocytes play important roles in the function and survival of neuronal cells. Dysfunctions of astrocytes are associated with numerous disorders and diseases of the nervous system, including motor neuron diseases such as amyotrophic lateral sclerosis (ALS). Human-induced pluripotent stem cell (iPSC)-based approaches are becoming increasingly important for the study of the mechanisms underlying the involvement of astrocytes in non-cell autonomous processes of motor neuron degeneration in ALS. These studies must account for the molecular and functional diversity among astrocytes in different regions of the brain and spinal cord. It is essential that the most pathologically relevant astrocyte preparations are used when investigating non-cell autonomous mechanisms of either upper or lower motor neuron degeneration in ALS. Here, we describe the efficient and streamlined generation of human iPSC-derived astrocytes with molecular and biological properties similar to physiological astrocytes in the ventral spinal cord. These induced astrocytes exhibit spontaneous and ATP-induced calcium transients, and lack signs of overt activation. Human iPSC-derived astrocytes with ventral spinal cord features offer advantages over more generic astrocyte preparations for the study of both ventral spinal cord astrocyte biology and the involvement of astrocytes in mechanisms of lower motor neuron degeneration in ALS.


Assuntos
Esclerose Lateral Amiotrófica , Células-Tronco Pluripotentes Induzidas , Esclerose Lateral Amiotrófica/patologia , Astrócitos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Neurônios Motores/patologia , Degeneração Neural/patologia
15.
Exp Cell Res ; 316(14): 2365-76, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20580711

RESUMO

Previous gene targeting studies in mice have implicated the nuclear protein Krüppel-like factor 7 (KLF7) in nervous system development while cell culture assays have documented its involvement in cell cycle regulation. By employing short hairpin RNA (shRNA)-mediated gene silencing, here we demonstrate that murine Klf7 gene expression is required for in vitro differentiation of neuroectodermal and mesodermal cells. Specifically, we show a correlation of Klf7 silencing with down-regulation of the neuronal marker microtubule-associated protein 2 (Map2) and the nerve growth factor (NGF) tyrosine kinase receptor A (TrkA) using the PC12 neuronal cell line. Similarly, KLF7 inactivation in Klf7-null mice decreases the expression of the neurogenic marker brain lipid-binding protein/fatty acid-binding protein 7 (BLBP/FABP7) in neural stem cells (NSCs). We also report that Klf7 silencing is detrimental to neuronal and cardiomyocytic differentiation of embryonic stem cells (ESCs), in addition to altering the adipogenic and osteogenic potential of mouse embryonic fibroblasts (MEFs). Finally, our results suggest that genes that are key for self-renewal of undifferentiated ESCs repress Klf7 expression in ESCs. Together with previous findings, these results provide evidence that KLF7 has a broad spectrum of regulatory functions, which reflect the discrete cellular and molecular contexts in which this transcription factor operates.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , Mesoderma/citologia , Placa Neural/citologia , Neurônios/citologia , Animais , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/citologia , Feminino , Inativação Gênica , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Células PC12 , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Proc Natl Acad Sci U S A ; 105(17): 6451-6, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18427115

RESUMO

Individual spinal motor neuron identities are specified in large part by the intrinsic repertoire of transcription factors expressed by undifferentiated progenitors and maturing neurons. It is shown here that the transcription factor AML1/Runx1 (Runx1) is expressed in selected spinal motor neuron subtypes after the onset of differentiation and is both necessary and sufficient to suppress interneuron-specific developmental programs and promote maintenance of motor neuron characteristics. These findings show an important role for Runx1 during the consolidation of selected spinal motor neuron identities. Moreover, they suggest a requirement for a persistent suppression of interneuron genes within maturing motor neurons.


Assuntos
Linhagem da Célula , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Interneurônios/metabolismo , Neurônios Motores/citologia , Medula Espinal/citologia , Animais , Diferenciação Celular , Sobrevivência Celular , Feminino , Interneurônios/citologia , Masculino , Camundongos , Mitose , Neurônios Motores/metabolismo , Medula Espinal/metabolismo
17.
Front Cell Neurosci ; 15: 707861, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34602979

RESUMO

Astrocytes are a large group of glial cells that perform a variety of physiological functions in the nervous system. They provide trophic, as well as structural, support to neuronal cells. Astrocytes are also involved in neuroinflammatory processes contributing to neuronal dysfunction and death. Growing evidence suggests important roles for astrocytes in non-cell autonomous mechanisms of motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Understanding these mechanisms necessitates the combined use of animal and human cell-based experimental model systems, at least in part because human astrocytes display a number of unique features that cannot be recapitulated in animal models. Human induced pluripotent stem cell (hiPSC)-based approaches provide the opportunity to generate disease-relevant human astrocytes to investigate the roles of these cells in ALS. These approaches are facing the growing recognition that there are heterogenous populations of astrocytes in the nervous system which are not functionally equivalent. This review will discuss the importance of taking astrocyte heterogeneity into consideration when designing hiPSC-based strategies aimed at generating the most informative preparations to study the contribution of astrocytes to ALS pathophysiology.

18.
Genes Brain Behav ; 20(1): e12686, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32691490

RESUMO

Understanding the rules that govern neuronal dynamics throughout the brain to subserve behavior and cognition remains one of the biggest challenges in neuroscience research. Recent technical advances enable the recording of increasingly larger neuronal populations to produce increasingly more sophisticated datasets. Despite bold and important open-science and data-sharing policies, these datasets tend to include unique data acquisition methods, behaviors, and file structures. Discrepancies between experimental protocols present key challenges in comparing data between laboratories and across different brain regions and species. Here, we discuss our recent efforts to create a standardized and high-throughput research platform to address these issues. The McGill-Mouse-Miniscope (M3) platform is an initiative to combine miniscope calcium imaging with standardized touchscreen-based animal behavioral testing. The goal is to curate an open-source and standardized framework for acquiring, analyzing, and accessing high-quality data of the neuronal dynamics that underly cognition throughout the brain in mice, marmosets, and models of disease. We end with a discussion of future developments and a call for users to adopt this standardized approach.


Assuntos
Pesquisa Comportamental/instrumentação , Encéfalo/fisiologia , Interface Usuário-Computador , Animais , Pesquisa Comportamental/métodos , Encéfalo/citologia , Encéfalo/metabolismo , Cálcio/metabolismo , Cognição , Ensaios de Triagem em Larga Escala/instrumentação , Ensaios de Triagem em Larga Escala/métodos , Camundongos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Neurônios/metabolismo , Neurônios/fisiologia
19.
J Cell Biol ; 220(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33443570

RESUMO

Glioblastoma is the most common and deadly malignant brain cancer. We now demonstrate that loss of function of the endosomal GTPase Rab35 in human brain tumor initiating cells (BTICs) increases glioblastoma growth and decreases animal survival following BTIC implantation in mouse brains. Mechanistically, we identify that the GTPase Arf5 interacts with the guanine nucleotide exchange factor (GEF) for Rab35, DENND1/connecdenn, and allosterically enhances its GEF activity toward Rab35. Knockdown of either Rab35 or Arf5 increases cell migration, invasiveness, and self-renewal in culture and enhances the growth and invasiveness of BTIC-initiated brain tumors in mice. RNAseq of the tumors reveals up-regulation of the tumor-promoting transcription factor SPOCD1, and disruption of the Arf5/Rab35 axis in glioblastoma cells leads to strong activation of the epidermal growth factor receptor, with resulting enhancement of SPOCD1 levels. These discoveries reveal an unexpected cascade between an Arf and a Rab and indicate a role for the cascade, and thus endosomal trafficking, in brain tumors.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteínas rab de Ligação ao GTP/metabolismo , Regulação Alostérica , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Autorrenovação Celular , Receptores ErbB/metabolismo , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Invasividade Neoplásica , Ligação Proteica , Domínios Proteicos , Transdução de Sinais , Análise de Sobrevida
20.
Nat Neurosci ; 9(6): 743-51, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16680166

RESUMO

In the developing cerebral cortex, neurons are born on a predictable schedule. Here we show in mice that the essential timing mechanism is programmed within individual progenitor cells, and its expression depends solely on cell-intrinsic and environmental factors generated within the clonal lineage. Multipotent progenitor cells undergo repeated asymmetric divisions, sequentially generating neurons in their normal in vivo order: first preplate cells, including Cajal-Retzius neurons, then deep and finally superficial cortical plate neurons. As each cortical layer arises, stem cells and neuroblasts become restricted from generating earlier-born neuron types. Growth as neurospheres or in co-culture with younger cells did not restore their plasticity. Using short-hairpin RNA (shRNA) to reduce Foxg1 expression reset the timing of mid- but not late-gestation progenitors, allowing them to remake preplate neurons and then cortical-plate neurons. Our data demonstrate that neural stem cells change neuropotency during development and have a window of plasticity when restrictions can be reversed.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Proliferação de Células , Córtex Cerebral/embriologia , Neurônios/metabolismo , Células-Tronco/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/genética , Divisão Celular/genética , Movimento Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Regulação para Baixo/genética , Proteínas da Matriz Extracelular/genética , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Microscopia de Vídeo , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal/genética , Neurônios/citologia , Interferência de RNA/fisiologia , Proteína Reelina , Serina Endopeptidases/genética , Células-Tronco/citologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa