Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell ; 186(7): 1295-1299, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-37001493

RESUMO

The transition to independence requires shared enthusiasm for one's research goals from broad audiences. In this commentary, we describe the use of "research vision workshopping" within peer mentoring networks. We contend that this approach is broadly useful for the development and refinement of research visions for the academic job search.


Assuntos
Tutoria , Humanos , Mentores , Grupo Associado
2.
Development ; 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38912572

RESUMO

The excitatory neurons of the three cerebellar nuclei (eCN) form the primary output for the cerebellum. The medial eCN (eCNm) were recently divided into molecularly defined subdomains in the adult, however how they are established during development is not known. We define molecular subdomains of the embryonic eCNm using scRNA-seq and spatial expression analysis, showing they evolve during embryogenesis to prefigure the adult. Furthermore, the medial eCN are transcriptionally divergent from the other nuclei by E14.5. We previously showed that loss of the homeobox genes En1 and En2 leads to loss of approximately half of embryonic eCNm. We demonstrate that mutation of En1/2 in embryonic eCNm results in death of specific posterior eCNm molecular subdomains and down regulation of TBR2 (EOMES) in an anterior embryonic subdomain, as well as reduced synaptic gene expression. We further reveal a similar function for EN1/2 in mediating TBR2 expression, neuron differentiation and survival in the other excitatory neurons (granule and unipolar brush cells). Thus, our work defines embryonic eCNm molecular diversity and reveals conserved roles for EN1/2 in the cerebellar excitatory neuron lineage.

3.
Development ; 149(18)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36172987

RESUMO

The cerebellum has a simple cytoarchitecture consisting of a folded cortex with three cell layers that surrounds a nuclear structure housing the output neurons. The excitatory neurons are generated from a unique progenitor zone, the rhombic lip, whereas the inhibitory neurons and astrocytes are generated from the ventricular zone. The growth phase of the cerebellum is driven by lineage-restricted progenitor populations derived from each zone. Research during the past decade has uncovered the importance of cell-to-cell communication between the lineages through largely unknown signaling mechanisms for regulating the scaling of cell numbers and cell plasticity during mouse development and following injury in the neonatal (P0-P14) cerebellum. This Review focuses on how the interplay between cell types is key to morphogenesis, production of robust neural circuits and replenishment of cells after injury, and ends with a discussion of the implications of the greater complexity of the human cerebellar progenitor zones for development and disease.


Assuntos
Cerebelo , Neurônios , Animais , Astrócitos , Humanos , Camundongos , Morfogênese , Neurônios/metabolismo , Células de Purkinje
4.
Neuroimage ; 276: 120198, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37245561

RESUMO

Magnetic Resonance Imaging (MRI) resolution continues to improve, making it important to understand the cellular basis for different MRI contrast mechanisms. Manganese-enhanced MRI (MEMRI) produces layer-specific contrast throughout the brain enabling in vivo visualization of cellular cytoarchitecture, particularly in the cerebellum. Due to the unique geometry of the cerebellum, especially near the midline, 2D MEMRI images can be acquired from a relatively thick slice by averaging through areas of uniform morphology and cytoarchitecture to produce very high-resolution visualization of sagittal planes. In such images, MEMRI hyperintensity is uniform in thickness throughout the anterior-posterior axis of sagittal sections and is centrally located in the cerebellar cortex. These signal features suggested that the Purkinje cell layer, which houses the cell bodies of the Purkinje cells and the Bergmann glia, is the source of hyperintensity. Despite this circumstantial evidence, the cellular source of MRI contrast has been difficult to define. In this study, we quantified the effects of selective ablation of Purkinje cells or Bergmann glia on cerebellar MEMRI signal to determine whether signal could be assigned to one cell type. We found that the Purkinje cells, not the Bergmann glia, are the primary of source of the enhancement in the Purkinje cell layer. This cell-ablation strategy should be useful for determining the cell specificity of other MRI contrast mechanisms.


Assuntos
Cerebelo , Manganês , Humanos , Manganês/metabolismo , Cerebelo/patologia , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Neuroglia/metabolismo , Imageamento por Ressonância Magnética/métodos
5.
Development ; 147(10)2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32366677

RESUMO

Thanks to many advances in genetic manipulation, mouse models have become very powerful in their ability to interrogate biological processes. In order to precisely target expression of a gene of interest to particular cell types, intersectional genetic approaches using two promoter/enhancers unique to a cell type are ideal. Within these methodologies, variants that add temporal control of gene expression are the most powerful. We describe the development, validation and application of an intersectional approach that involves three transgenes, requiring the intersection of two promoter/enhancers to target gene expression to precise cell types. Furthermore, the approach uses available lines expressing tTA/rTA to control the timing of gene expression based on whether doxycycline is absent or present, respectively. We also show that the approach can be extended to other animal models, using chicken embryos. We generated three mouse lines targeted at the Tigre (Igs7) locus with TRE-loxP-tdTomato-loxP upstream of three genes (p21, DTA and Ctgf), and combined them with Cre and tTA/rtTA lines that target expression to the cerebellum and limbs. Our tools will facilitate unraveling biological questions in multiple fields and organisms.


Assuntos
Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Transgenes , Animais , Cerebelo , Embrião de Galinha , Doxiciclina/farmacologia , Extremidades , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Transativadores/genética , Transcrição Gênica
6.
EMBO J ; 31(15): 3351-62, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22713866

RESUMO

The anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase is tightly regulated to ensure programmed proteolysis in cells. The activity of the APC/C is positively controlled by cyclin-dependent kinase (CDK), but a second level of control must also exist because phosphorylation inactivates Cdc20, a mitotic APC/C co-activator. How Cdc20 is dephosphorylated specifically, when CDK is high, has remained unexplained. Here, we show that phosphatases are crucial to activate the APC/C. Cdc20 is phosphorylated at six conserved residues (S50/T64/T68/T79/S114/S165) by CDK in Xenopus egg extracts. When all the threonine residues are phosphorylated, Cdc20 binding to and activation of the APC/C are inhibited. Their dephosphorylation is regulated depending on the sites and protein phosphatase 2A, active in mitosis, is essential to dephosphorylate the threonine residues and activate the APC/C. Consistently, most of the Cdc20 bound to the APC/C in anaphase evades phosphorylation at T79. Furthermore, we show that the 'activation domain' of Cdc20 associates with the Apc6 and Apc8 core subunits. Our data suggest that dephosphorylation of Cdc20 is required for its loading and activation of the APC/C ubiquitin ligase.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteínas de Xenopus/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteínas Cdc20 , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiologia , Células Cultivadas , Ativação Enzimática , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Mitose/genética , Mitose/fisiologia , Modelos Biológicos , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional/genética , Estrutura Terciária de Proteína/fisiologia , Spodoptera , Complexos Ubiquitina-Proteína Ligase/química , Proteínas de Xenopus/química , Proteínas de Xenopus/fisiologia , Xenopus laevis
7.
Sci Adv ; 10(12): eadn4649, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38517960

RESUMO

Genomic rearrangements are a hallmark of most childhood tumors, including medulloblastoma, one of the most common brain tumors in children, but their causes remain largely unknown. Here, we show that PiggyBac transposable element derived 5 (Pgbd5) promotes tumor development in multiple developmentally accurate mouse models of Sonic Hedgehog (SHH) medulloblastoma. Most Pgbd5-deficient mice do not develop tumors, while maintaining normal cerebellar development. Ectopic activation of SHH signaling is sufficient to enforce cerebellar granule cell progenitor-like cell states, which exhibit Pgbd5-dependent expression of distinct DNA repair and neurodevelopmental factors. Mouse medulloblastomas expressing Pgbd5 have increased numbers of somatic structural DNA rearrangements, some of which carry PGBD5-specific sequences at their breakpoints. Similar sequence breakpoints recurrently affect somatic DNA rearrangements of known tumor suppressors and oncogenes in medulloblastomas in 329 children. This identifies PGBD5 as a medulloblastoma mutator and provides a genetic mechanism for the generation of oncogenic DNA rearrangements in childhood cancer.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Humanos , Criança , Animais , Camundongos , Meduloblastoma/genética , Transposases/genética , Transposases/metabolismo , Proteínas Hedgehog/metabolismo , Fatores de Transcrição/genética , Mutagênese , Neoplasias Cerebelares/genética
8.
bioRxiv ; 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38077070

RESUMO

The excitatory neurons of the three cerebellar nuclei (eCN) form the primary output for the cerebellar circuit. The medial eCN (eCNm) were recently divided into molecularly defined subdomains in the adult, however how they are established during development is not known. We define molecular subdomains of the eCNm using scRNA-seq and spatial expression analysis and show they evolve during embryogenesis to resemble the adult. Furthermore, the eCNm is transcriptionally divergent from the rest of the eCN by E14.5. We previously showed that loss of the homeobox genes En1 and En2 leads to death of a subset of embryonic eCNm. We demonstrate that mutation of En1/2 in embryonic eCNm results in cell death of specific posterior eCNm molecular subdomains and loss of TBR2 (EOMES) expression in an anterior subdomain, as well as reduced synaptic gene expression. We further reveal a similar function for EN1/2 in mediating TBR2 expression, neuron differentiation and survival in the two other cerebellar excitatory neuron types. Thus, our work defines embryonic eCNm molecular diversity and reveals conserved roles for EN1/2 in the cerebellar excitatory neuron lineage.

9.
iScience ; 26(10): 107831, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37822508

RESUMO

The major cause of treatment failure and mortality among medulloblastoma patients is metastasis intracranially or along the spinal cord. The molecular mechanisms driving tumor metastasis in Sonic hedgehog-driven medulloblastoma (SHH-MB) patients, however, remain largely unknown. In this study we define a tumor suppressive role of KMT2D (MLL2), a gene frequently mutated in the most metastatic ß-subtype. Strikingly, genetic mouse models of SHH-MB demonstrate that heterozygous loss of Kmt2d in conjunction with activation of the SHH pathway causes highly penetrant disease with decreased survival, increased hindbrain invasion and spinal cord metastasis. Loss of Kmt2d attenuates neural differentiation and shifts the transcriptional/chromatin landscape of primary and metastatic tumors toward a decrease in differentiation genes and tumor suppressors and an increase in genes/pathways implicated in advanced stage cancer and metastasis (TGFß, Notch, Atoh1, Sox2, and Myc). Thus, secondary heterozygous KMT2D mutations likely have prognostic value for identifying SHH-MB patients prone to develop metastasis.

10.
Sci Adv ; 7(50): eabj1598, 2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34878841

RESUMO

To understand repair processes, it is critical to identify the molecular foundations underlying progenitor diversity and plasticity. Upon injury to the neonatal cerebellum, a normally gliogenic nestin-expressing progenitor (NEP) in the Bergmann glia layer (BgL) undergoes adaptive reprograming to restore granule cell production. However, the cellular states and genes regulating the NEP fate switch are unknown. Using single-cell RNA sequencing and fate mapping, we defined molecular subtypes of NEPs and their lineages under homeostasis and repair. NEPs contain two major subtypes: Hopx+ astrogliogenic and Ascl1+ neurogenic NEPs that are further subdivided based on their location, lineage, and differentiation status. Upon injury, an Ascl1+ transitory cellular state arises from Hopx+ BgL-NEPs. Furthermore, mutational analysis revealed that induction of Ascl1 is required for adaptive reprogramming by orchestrating a glial-to-neural switch in vivo following injury. Thus, we provide molecular and cellular insights into context-dependent progenitor plasticity and repair mechanisms in the brain.

11.
Cell Rep ; 31(12): 107805, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32579931

RESUMO

In the adult ventricular-subventricular zone (V-SVZ), neural stem cells (NSCs) generate new olfactory bulb (OB) neurons and glia throughout life. To map adult neuronal lineage progression, we profiled >56,000 V-SVZ and OB cells by single-cell RNA sequencing (scRNA-seq). Our analyses reveal the molecular diversity of OB neurons, including fate-mapped neurons, lineage progression dynamics, and an NSC intermediate enriched for Notum, which encodes a secreted WNT antagonist. SCOPE-seq technology, which links live-cell imaging with scRNA-seq, uncovers cell-size transitions during NSC differentiation and preferential NOTUM binding to proliferating neuronal precursors. Consistently, application of NOTUM protein in slice cultures and pharmacological inhibition of NOTUM in slice cultures and in vivo demonstrated that NOTUM negatively regulates V-SVZ proliferation. Timely, context-dependent neurogenesis demands adaptive signaling among neighboring progenitors. Our findings highlight a critical regulatory state during NSC activation marked by NOTUM, which attenuates WNT-stimulated proliferation in NSC progeny.


Assuntos
Envelhecimento/metabolismo , Linhagem da Célula , Esterases/metabolismo , Ventrículos Laterais/citologia , Neurogênese , Análise de Célula Única , Animais , Proliferação de Células , Regulação da Expressão Gênica , Genes Reporter , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Bulbo Olfatório/citologia
12.
Elife ; 82019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31742552

RESUMO

For neural systems to function effectively, the numbers of each cell type must be proportioned properly during development. We found that conditional knockout of the mouse homeobox genes En1 and En2 in the excitatory cerebellar nuclei neurons (eCN) leads to reduced postnatal growth of the cerebellar cortex. A subset of medial and intermediate eCN are lost in the mutants, with an associated cell non-autonomous loss of their presynaptic partner Purkinje cells by birth leading to proportional scaling down of neuron production in the postnatal cerebellar cortex. Genetic killing of embryonic eCN throughout the cerebellum also leads to loss of Purkinje cells and reduced postnatal growth but throughout the cerebellar cortex. Thus, the eCN play a key role in scaling the size of the cerebellum by influencing the survival of their Purkinje cell partners, which in turn regulate production of granule cells and interneurons via the amount of sonic hedgehog secreted.


Assuntos
Proliferação de Células , Córtex Cerebelar/crescimento & desenvolvimento , Núcleos Cerebelares/citologia , Células de Purkinje/fisiologia , Animais , Técnicas de Inativação de Genes , Proteínas de Homeodomínio/genética , Camundongos , Proteínas do Tecido Nervoso/deficiência
13.
Methods Mol Biol ; 1741: 91-101, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29392693

RESUMO

Several lines of evidence suggest a cellular hierarchy in glioblastoma (GBM). In this hierarchy, GBM stem-like cells (GSCs) play critical roles in tumor progression and recurrence, by virtue of their robust tumor-propagating potential and resistance to conventional chemoradiotherapy. Therefore, targeting GSCs holds significant therapeutic promise. Expression of CD133 (PROM1), a cell surface glycoprotein, has been associated with the GSC phenotype and used as a GSC marker. Here, we describe a protocol that allows the selective lentiviral transduction of CD133-expressing GBM cells. This selectivity is conferred by pseudotyping the lentiviral envelope with a single-chain antibody against an extracellular epitope on CD133. We previously demonstrated the efficacy and specificity of this lentiviral vector using patient-derived GBM cultures. This chapter outlines the preparation of the vector and the transduction of human GBM cells.


Assuntos
Antígeno AC133/genética , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Lentivirus/genética , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Expressão Gênica , Glioblastoma/terapia , Células HEK293 , Humanos , Camundongos , Células-Tronco Neoplásicas/patologia , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Methods Mol Biol ; 1741: 199-207, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29392702

RESUMO

Histologic heterogeneity in glioblastoma (GBM) is highlighted by regional variability in vascular density. Areas of vascular hyperplasia are interspersed with avascular territories, in which necrosis is surrounded by a zone of hypoxic tumor cells expressing stem cell markers, a phenomenon known as pseudopalisading necrosis. This vascular heterogeneity suggests intratumoral oxygen gradients, which regulate cellular and metabolic adaptations in tumor cells. Quantification of tumor vascularity, blood perfusion and oxygenation is therefore critical. In this chapter, we describe three different methods, all of which involve microscopy to analyze these parameters in tumor xenografts. We present detailed protocols for analysis of tumor endothelium using endothelial markers, blood perfusion by systemic infusion of Evans Blue and oxygen tension by pimonidazole injection, followed by immunostaining.


Assuntos
Xenoenxertos , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Oxigênio/metabolismo , Animais , Modelos Animais de Doenças , Imunofluorescência , Humanos , Hipóxia/metabolismo , Camundongos , Microscopia de Fluorescência , Estadiamento de Neoplasias , Nitroimidazóis/farmacologia
15.
Methods Mol Biol ; 1741: 171-182, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29392699

RESUMO

Glioblastoma (GBM), a deadly primary brain malignancy, manifests pronounced radioresistance. Identifying agents that improve the sensitivity of tumor tissue to radiotherapy is critical for improving patient outcomes. The response to ionizing radiation is regulated by both cell-intrinsic and -extrinsic mechanisms. In particular, the tumor microenvironment is known to promote radioresistance in GBM. Therefore, model systems used to test radiosensitizing agents need to take into account the tumor microenvironment. We recently showed that GBM explant cultures represent an adaptable ex vivo platform for rapid and personalized testing of radiosensitizers. These explants preserve the cellular composition and tissue architecture of parental patient tumors and therefore capture the microenvironmental context that critically determines the response to radiotherapy. This chapter focuses on the detailed protocol for testing candidate radiosensitizing agents in GBM explants.


Assuntos
Glioblastoma/patologia , Técnicas de Cultura de Órgãos , Tolerância a Radiação , Radiossensibilizantes/farmacologia , Imunofluorescência , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Humanos , Processamento de Imagem Assistida por Computador , Microscopia de Fluorescência , Tolerância a Radiação/efeitos dos fármacos , Radiação Ionizante , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos da radiação
16.
Elife ; 72018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30091706

RESUMO

Outside of the neurogenic niches of the brain, postmitotic neurons have not been found to undergo efficient regeneration. We demonstrate that mouse Purkinje cells (PCs), which are born at midgestation and are crucial for development and function of cerebellar circuits, are rapidly and fully regenerated following their ablation at birth. New PCs are produced from immature FOXP2+ Purkinje cell precursors (iPCs) that are able to enter the cell cycle and support normal cerebellum development. The number of iPCs and their regenerative capacity, however, diminish soon after birth and consequently PCs are poorly replenished when ablated at postnatal day five. Nevertheless, the PC-depleted cerebella reach a normal size by increasing cell size, but scaling of neuron types is disrupted and cerebellar function is impaired. Our findings provide a new paradigm in the field of neuron regeneration by identifying a population of immature neurons that buffers against perinatal brain injury in a stage-dependent process.


Assuntos
Proliferação de Células , Cerebelo/crescimento & desenvolvimento , Cerebelo/lesões , Células de Purkinje/fisiologia , Regeneração , Células-Tronco/fisiologia , Fatores Etários , Animais , Camundongos
17.
Methods Mol Biol ; 1741: 151-170, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29392698

RESUMO

Single-cell RNA sequencing (sc-RNASeq) is a recently developed technique used to evaluate the transcriptome of individual cells. As opposed to conventional RNASeq in which entire populations are sequenced in bulk, sc-RNASeq can be beneficial when trying to better understand gene expression patterns in markedly heterogeneous populations of cells or when trying to identify transcriptional signatures of rare cells that may be underrepresented when using conventional bulk RNASeq. In this method, we describe the generation and analysis of cDNA libraries from single patient-derived glioblastoma cells using the C1 Fluidigm system. The protocol details the use of the C1 integrated fluidics circuit (IFC) for capturing, imaging and lysing cells; performing reverse transcription; and generating cDNA libraries that are ready for sequencing and analysis.


Assuntos
Glioblastoma/genética , Sequenciamento de Nucleotídeos em Larga Escala , Análise de Sequência de RNA , Análise de Célula Única , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Biblioteca Gênica , Humanos , Técnicas de Amplificação de Ácido Nucleico , Análise de Célula Única/métodos , Transcriptoma
18.
Sci Adv ; 4(10): eaau5935, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30402543

RESUMO

A methionine substitution at lysine-27 on histone H3 variants (H3K27M) characterizes ~80% of diffuse intrinsic pontine gliomas (DIPG) and inhibits polycomb repressive complex 2 (PRC2) in a dominant-negative fashion. Yet, the mechanisms for this inhibition and abnormal epigenomic landscape have not been resolved. Using quantitative proteomics, we discovered that robust PRC2 inhibition requires levels of H3K27M greatly exceeding those of PRC2, seen in DIPG. While PRC2 inhibition requires interaction with H3K27M, we found that this interaction on chromatin is transient, with PRC2 largely being released from H3K27M. Unexpectedly, inhibition persisted even after PRC2 dissociated from H3K27M-containing chromatin, suggesting a lasting impact on PRC2. Furthermore, allosterically activated PRC2 is particularly sensitive to H3K27M, leading to the failure to spread H3K27me from PRC2 recruitment sites and consequently abrogating PRC2's ability to establish H3K27me2-3 repressive chromatin domains. In turn, levels of polycomb antagonists such as H3K36me2 are elevated, suggesting a more global, downstream effect on the epigenome. Together, these findings reveal the conditions required for H3K27M-mediated PRC2 inhibition and reconcile seemingly paradoxical effects of H3K27M on PRC2 recruitment and activity.


Assuntos
Neoplasias do Tronco Encefálico/patologia , Cromatina/química , Glioma/patologia , Histonas/metabolismo , Lisina/metabolismo , Complexo Repressor Polycomb 2/antagonistas & inibidores , Animais , Neoplasias do Tronco Encefálico/genética , Neoplasias do Tronco Encefálico/metabolismo , Células Cultivadas , Criança , Cromatina/genética , Cromatina/metabolismo , Modelos Animais de Doenças , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo
19.
Nat Neurosci ; 20(10): 1361-1370, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28805814

RESUMO

Regeneration of several organs involves adaptive reprogramming of progenitors, but the intrinsic capacity of the developing brain to replenish lost cells remains largely unknown. Here we found that the developing cerebellum has unappreciated progenitor plasticity, since it undergoes near full growth and functional recovery following acute depletion of granule cells, the most plentiful neuron population in the brain. We demonstrate that following postnatal ablation of granule cell progenitors, Nestin-expressing progenitors, specified during mid-embryogenesis to produce astroglia and interneurons, switch their fate and generate granule neurons in mice. Moreover, Hedgehog signaling in two Nestin-expressing progenitor populations is crucial not only for the compensatory replenishment of granule neurons but also for scaling interneuron and astrocyte numbers. Thus, we provide insights into the mechanisms underlying robustness of circuit formation in the cerebellum and speculate that adaptive reprogramming of progenitors in other brain regions plays a greater role than appreciated in developmental regeneration.


Assuntos
Cerebelo/fisiologia , Nestina/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/fisiologia , Cerebelo/efeitos da radiação , Feminino , Proteínas Hedgehog/fisiologia , Interneurônios/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Nestina/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/efeitos da radiação
20.
Oncotarget ; 8(39): 64932-64953, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-29029402

RESUMO

Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA