Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Neuron ; 110(23): 3936-3951.e10, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36174572

RESUMEN

Zika virus (ZIKV) can infect human developing brain (HDB) progenitors resulting in epidemic microcephaly, whereas analogous cellular tropism offers treatment potential for the adult brain cancer, glioblastoma (GBM). We compared productive ZIKV infection in HDB and GBM primary tissue explants that both contain SOX2+ neural progenitors. Strikingly, although the HDB proved uniformly vulnerable to ZIKV infection, GBM was more refractory, and this correlated with an innate immune expression signature. Indeed, GBM-derived CD11b+ microglia/macrophages were necessary and sufficient to protect progenitors against ZIKV infection in a non-cell autonomous manner. Using SOX2+ GBM cell lines, we found that CD11b+-conditioned medium containing type 1 interferon beta (IFNß) promoted progenitor resistance to ZIKV, whereas inhibition of JAK1/2 signaling restored productive infection. Additionally, CD11b+ conditioned medium, and IFNß treatment rendered HDB progenitor lines and explants refractory to ZIKV. These findings provide insight into neuroprotection for HDB progenitors as well as enhanced GBM oncolytic therapies.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Humanos , Células Mieloides , Células Madre , Interferones
2.
Development ; 149(20)2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35748297

RESUMEN

Oligodendrocytes, the myelinating cells of the central nervous system, possess great potential for disease modeling and cell transplantation-based therapies for leukodystrophies. However, caveats to oligodendrocyte differentiation protocols ( Ehrlich et al., 2017; Wang et al., 2013; Douvaras and Fossati, 2015) from human embryonic stem and induced pluripotent stem cells (iPSCs), which include slow and inefficient differentiation, and tumorigenic potential of contaminating undifferentiated pluripotent cells, are major bottlenecks towards their translational utility. Here, we report the rapid generation of human oligodendrocytes by direct lineage conversion of human dermal fibroblasts (HDFs). We show that the combination of the four transcription factors OLIG2, SOX10, ASCL1 and NKX2.2 is sufficient to convert HDFs to induced oligodendrocyte precursor cells (iOPCs). iOPCs resemble human primary and iPSC-derived OPCs based on morphology and transcriptomic analysis. Importantly, iOPCs can differentiate into mature myelinating oligodendrocytes in vitro and in vivo. Finally, iOPCs derived from patients with Pelizaeus Merzbacher disease, a hypomyelinating leukodystrophy caused by mutations in the proteolipid protein 1 (PLP1) gene, showed increased cell death compared with iOPCs from healthy donors. Thus, human iOPCs generated by direct lineage conversion represent an attractive new source for human cell-based disease models and potentially myelinating cell grafts.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Pelizaeus-Merzbacher , Diferenciación Celular/fisiología , Fibroblastos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Oligodendroglía/metabolismo , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/metabolismo , Enfermedad de Pelizaeus-Merzbacher/terapia
3.
Pediatr Res ; 90(6): 1161-1170, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33654279

RESUMEN

BACKGROUND: Neonatal stroke affects 1 in 2800 live births and is a major cause of neurological injury. The Sonic hedgehog (Shh) signaling pathway is critical for central nervous system (CNS) development and has neuroprotective and reparative effects in different CNS injury models. Previous studies have demonstrated beneficial effects of small molecule Shh-Smoothened agonist (SAG) against neonatal cerebellar injury and it improves Down syndrome-related brain structural deficits in mice. Here we investigated SAG neuroprotection in rat models of neonatal ischemia-reperfusion (stroke) and adult focal white matter injury. METHODS: We used transient middle cerebral artery occlusion at P10 and ethidium bromide (EB) injection in adult rats to induce damage. Following surgery and SAG or vehicle treatment, we analyzed tissue loss, cell proliferation and fate, and behavioral outcome. RESULTS: We report that a single dose of SAG administered following neonatal stroke preserved brain volume, reduced gliosis, enhanced oligodendrocyte progenitor cell (OPC) and EC proliferation, and resulted in long-term cognitive improvement. Single-dose SAG also promoted proliferation of OPCs following focal demyelination in the adult rat. CONCLUSIONS: These findings indicate benefit of one-time SAG treatment post insult in reducing brain injury and improving behavioral outcome after experimental neonatal stroke. IMPACT: A one-time dose of small molecule Sonic hedgehog agonist protected against neonatal stroke and improved long-term behavioral outcomes in a rat model. This study extends the use of Sonic hedgehog in treating developing brain injury, previously shown in animal models of Down syndrome and cerebellar injury. Sonic hedgehog agonist is one of the most promising therapies in treating neonatal stroke thanks to its safety profile and low dosage.


Asunto(s)
Proteínas Hedgehog/antagonistas & inhibidores , Fármacos Neuroprotectores/uso terapéutico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Accidente Cerebrovascular/prevención & control , Animales , Conducta Animal , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Recién Nacido , Infarto de la Arteria Cerebral Media/complicaciones , Ratones , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/etiología
4.
Cell ; 182(3): 594-608.e11, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32679030

RESUMEN

Human cerebral cortex size and complexity has increased greatly during evolution. While increased progenitor diversity and enhanced proliferative potential play important roles in human neurogenesis and gray matter expansion, the mechanisms of human oligodendrogenesis and white matter expansion remain largely unknown. Here, we identify EGFR-expressing "Pre-OPCs" that originate from outer radial glial cells (oRGs) and undergo mitotic somal translocation (MST) during division. oRG-derived Pre-OPCs provide an additional source of human cortical oligodendrocyte precursor cells (OPCs) and define a lineage trajectory. We further show that human OPCs undergo consecutive symmetric divisions to exponentially increase the progenitor pool size. Additionally, we find that the OPC-enriched gene, PCDH15, mediates daughter cell repulsion and facilitates proliferation. These findings indicate properties of OPC derivation, proliferation, and dispersion important for human white matter expansion and myelination.


Asunto(s)
Cadherinas/metabolismo , Corteza Cerebral/citología , Células Ependimogliales/metabolismo , Neurogénesis/genética , Células Precursoras de Oligodendrocitos/metabolismo , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Proliferación Celular/genética , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Células Ependimogliales/citología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Células Precursoras de Oligodendrocitos/citología , ARN Interferente Pequeño , RNA-Seq , Análisis de la Célula Individual , Sustancia Blanca/citología , Sustancia Blanca/embriología , Sustancia Blanca/metabolismo
5.
Phys Chem Chem Phys ; 22(26): 14976-14982, 2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32588846

RESUMEN

Machine learning is a valuable tool in the development of chemical technologies but its applications into supramolecular chemistry have been limited. Here, the utility of kernel-based support vector machine learning using density functional theory calculations as training data is evaluated when used to predict equilibrium binding coefficients of small molecules with cucurbit[7]uril (CB[7]). We find that utilising SVMs may confer some predictive ability. This algorithm was then used to predict the binding of drugs TAK-580 and selumetinib. The algorithm did predict strong binding for TAK-580 and poor binding for selumetinib, and these results were experimentally validated. It was discovered that the larger homologue cucurbit[8]uril (CB[8]) is partial to selumetinib, suggesting an opportunity for tunable release by introducing different concentrations of CB[7] or CB[8] into a hydrogel depot. We qualitatively demonstrated that these drugs may have utility in combination against gliomas. Finally, mass transfer simulations show CB[7] can independently tune the release of TAK-580 without affecting selumetinib. This work gives specific evidence that a machine learning approach to recognition of small molecules by macrocycles has merit and reinforces the view that machine learning may prove valuable in the development of drug delivery systems and supramolecular chemistry more broadly.


Asunto(s)
Bencimidazoles/química , Hidrocarburos Aromáticos con Puentes/química , Compuestos Heterocíclicos con 3 Anillos/química , Imidazoles/química , Teoría Funcional de la Densidad , Modelos Químicos , Máquina de Vectores de Soporte
6.
Neuron ; 106(4): 607-623.e5, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32183943

RESUMEN

Postnatal brain circuit assembly is driven by temporally regulated intrinsic and cell-extrinsic cues that organize neurogenesis, migration, and axo-dendritic specification in post-mitotic neurons. While cell polarity is an intrinsic organizer of morphogenic events, environmental cues in the germinal zone (GZ) instructing neuron polarization and their coupling during postnatal development are unclear. We report that oxygen tension, which rises at birth, and the von Hippel-Lindau (VHL)-hypoxia-inducible factor 1α (Hif1α) pathway regulate polarization and maturation of post-mitotic cerebellar granule neurons (CGNs). At early postnatal stages with low GZ vascularization, Hif1α restrains CGN-progenitor cell-cycle exit. Unexpectedly, cell-intrinsic VHL-Hif1α pathway activation also delays the timing of CGN differentiation, germinal zone exit, and migration initiation through transcriptional repression of the partitioning-defective (Pard) complex. As vascularization proceeds, these inhibitory mechanisms are downregulated, implicating increasing oxygen tension as a critical switch for neuronal polarization and cerebellar GZ exit.


Asunto(s)
Polaridad Celular/fisiología , Cerebelo/crecimiento & desarrollo , Cerebelo/fisiología , Neurogénesis/fisiología , Neuronas/citología , Animales , Diferenciación Celular/fisiología , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Oxígeno , Transducción de Señal/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
7.
Cell Stem Cell ; 25(4): 531-541.e6, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585094

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.


Asunto(s)
Deferiprona/uso terapéutico , Células Madre Pluripotentes Inducidas/fisiología , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Proteína Proteolipídica de la Mielina/metabolismo , Oligodendroglía/fisiología , Enfermedad de Pelizaeus-Merzbacher/terapia , Animales , Diferenciación Celular , Células Cultivadas , Ferroptosis , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Peroxidación de Lípido , Ratones , Ratones Mutantes , Mutación/genética , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/efectos de los fármacos , Oligodendroglía/trasplante , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/patología , Trasplante de Células Madre , Reparación del Gen Blanco
8.
Stem Cell Reports ; 13(2): 254-261, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31378671

RESUMEN

Four boys with Pelizaeus-Merzbacher disease, an X-linked leukodystrophy, underwent transplantation with human allogeneic central nervous system stem cells (HuCNS-SC). Subsequently, all subjects were followed for an additional 4 years in this separate follow-up study to evaluate safety, neurologic function, magnetic resonance imaging (MRI) data, and immunologic response. The neurosurgical procedure, immunosuppression, and HuCNS-SC transplantation were well tolerated and all four subjects were alive at the conclusion of the study period. At year 2, all subjects exhibited diffusion MRI changes at the implantation sites as well as in more distant brain regions. There were persistent, increased signal changes in the three patients who were studied up to year 5. Two of four subjects developed donor-specific HLA alloantibodies, demonstrating that neural stem cells can elicit an immune response when injected into the CNS, and suggesting the importance of monitoring immunologic parameters and identifying markers of engraftment in future studies.


Asunto(s)
Encéfalo/diagnóstico por imagen , Células-Madre Neurales/trasplante , Enfermedad de Pelizaeus-Merzbacher/terapia , Encéfalo/fisiología , Preescolar , Estudios de Seguimiento , Antígenos HLA/inmunología , Humanos , Lactante , Isoanticuerpos/sangre , Imagen por Resonancia Magnética , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Enfermedad de Pelizaeus-Merzbacher/inmunología , Enfermedad de Pelizaeus-Merzbacher/patología , Índice de Severidad de la Enfermedad , Trasplante de Células Madre/efectos adversos , Trasplante Homólogo , Resultado del Tratamiento
9.
Nature ; 573(7772): 75-82, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31316211

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical grey versus subcortical white matter and neurodegeneration at chronic stages. Here we used single-nucleus RNA sequencing to assess changes in expression in multiple cell lineages in MS lesions and validated the results using multiplex in situ hybridization. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper-cortical layers underlying meningeal inflammation; such MS neuron populations exhibited upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated microglia mapped most strongly to the rim of MS plaques. Notably, single-nucleus RNA sequencing identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to progression of MS lesions.


Asunto(s)
Linaje de la Célula , Esclerosis Múltiple/patología , Neuronas/patología , Adulto , Animales , Astrocitos/metabolismo , Astrocitos/patología , Autopsia , Criopreservación , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Esclerosis Múltiple/genética , Vaina de Mielina/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Fagocitosis , ARN Nuclear Pequeño/análisis , ARN Nuclear Pequeño/genética , RNA-Seq , Transcriptoma/genética
11.
Nat Rev Clin Oncol ; 16(8): 509-520, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30733593

RESUMEN

Despite decades of research, brain tumours remain among the deadliest of all forms of cancer. The ability of these tumours to resist almost all conventional and novel treatments relates, in part, to the unique cell-intrinsic and microenvironmental properties of neural tissues. In an attempt to encourage progress in our understanding and ability to successfully treat patients with brain tumours, Cancer Research UK convened an international panel of clinicians and laboratory-based scientists to identify challenges that must be overcome if we are to cure all patients with a brain tumour. The seven key challenges summarized in this Position Paper are intended to serve as foci for future research and investment.


Asunto(s)
Neoplasias Encefálicas/terapia , Humanos
12.
Cancer Cell ; 33(5): 874-889.e7, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29681511

RESUMEN

Gliomas comprise heterogeneous malignant glial and stromal cells. While blood vessel co-option is a potential mechanism to escape anti-angiogenic therapy, the relevance of glial phenotype in this process is unclear. We show that Olig2+ oligodendrocyte precursor-like glioma cells invade by single-cell vessel co-option and preserve the blood-brain barrier (BBB). Conversely, Olig2-negative glioma cells form dense perivascular collections and promote angiogenesis and BBB breakdown, leading to innate immune cell activation. Experimentally, Olig2 promotes Wnt7b expression, a finding that correlates in human glioma profiling. Targeted Wnt7a/7b deletion or pharmacologic Wnt inhibition blocks Olig2+ glioma single-cell vessel co-option and enhances responses to temozolomide. Finally, Olig2 and Wnt7 become upregulated after anti-VEGF treatment in preclinical models and patients. Thus, glial-encoded pathways regulate distinct glioma-vascular microenvironmental interactions.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Glioma/irrigación sanguínea , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/microbiología , Proteínas Wnt/metabolismo , Animales , Bevacizumab/farmacología , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Ratones , Trasplante de Neoplasias , Factor de Transcripción 2 de los Oligodendrocitos/genética , Temozolomida/farmacología , Células Tumorales Cultivadas , Microambiente Tumoral , Proteínas Wnt/genética , Vía de Señalización Wnt/efectos de los fármacos
13.
Cerebellum ; 17(2): 213-227, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29134361

RESUMEN

The cerebellum undergoes rapid growth during the third trimester and is vulnerable to injury and deficient growth in infants born prematurely. Factors associated with preterm cerebellar hypoplasia include chronic lung disease and postnatal glucocorticoid administration. We modeled chronic hypoxemia and glucocorticoid administration in neonatal mice to study whole cerebellar and cell type-specific effects of dual exposure. Chronic neonatal hypoxia resulted in permanent cerebellar hypoplasia. This was compounded by administration of prednisolone as shown by greater volume loss and Purkinje cell death. In the setting of hypoxia and prednisolone, administration of a small molecule Smoothened-Hedgehog agonist (SAG) preserved cerebellar volume and protected against Purkinje cell death. Such protective effects were observed even when SAG was given as a one-time dose after dual insult. To model complex injury and determine cell type-specific roles for the hypoxia inducible factor (HIF) pathway, we performed conditional knockout of von Hippel Lindau (VHL) to hyperactivate HIF1α in cerebellar granule neuron precursors (CGNP) or Purkinje cells. Surprisingly, HIF activation in either cell type resulted in no cerebellar deficit. However, in mice administered prednisolone, HIF overactivation in CGNPs resulted in significant cerebellar hypoplasia, whereas HIF overactivation in Purkinje cells caused cell death. Together, these findings indicate that HIF primes both cell types for injury via glucocorticoids, and that hypoxia/HIF + postnatal glucocorticoid administration act on distinct cellular pathways to cause cerebellar injury. They further suggest that SAG is neuroprotective in the setting of complex neonatal cerebellar injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Cerebelo/anomalías , Ciclohexilaminas/uso terapéutico , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Tiofenos/uso terapéutico , Aminoácidos Dicarboxílicos/farmacología , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Cerebelo/efectos de los fármacos , Discapacidades del Desarrollo/etiología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Glucocorticoides/farmacología , Hipoxia Encefálica/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/etiología , Prednisolona/uso terapéutico , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
14.
Cereb Cortex ; 28(11): 3797-3815, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29028947

RESUMEN

The postnatal functions of the Dlx1&2 transcription factors in cortical interneurons (CINs) are unknown. Here, using conditional Dlx1, Dlx2, and Dlx1&2 knockouts (CKOs), we defined their roles in specific CINs. The CKOs had dendritic, synaptic, and survival defects, affecting even PV+ CINs. We provide evidence that DLX2 directly drives Gad1, Gad2, and Vgat expression, and show that mutants had reduced mIPSC amplitude. In addition, the mutants formed fewer GABAergic synapses on excitatory neurons and had reduced mIPSC frequency. Furthermore, Dlx1/2 CKO had hypoplastic dendrites, fewer excitatory synapses, and reduced excitatory input. We provide evidence that some of these phenotypes were due to reduced expression of GRIN2B (a subunit of the NMDA receptor), a high confidence Autism gene. Thus, Dlx1&2 coordinate key components of CIN postnatal development by promoting their excitability, inhibitory output, and survival.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Neuronas GABAérgicas/fisiología , Proteínas de Homeodominio/fisiología , Interneuronas/fisiología , Sinapsis/fisiología , Factores de Transcripción/fisiología , Ácido gamma-Aminobutírico/biosíntesis , Animales , Corteza Cerebral/citología , Femenino , Neuronas GABAérgicas/citología , Regulación del Desarrollo de la Expresión Génica , Glutamato Descarboxilasa/metabolismo , Proteínas de Homeodominio/genética , Interneuronas/citología , Masculino , Ratones Noqueados , Potenciales Postsinápticos Miniatura , Factores de Transcripción/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
15.
Cell Rep ; 18(13): 3167-3177, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28355568

RESUMEN

During development of the vertebrate CNS, the basic helix-loop-helix (bHLH) transcription factor Olig2 sustains replication competence of progenitor cells that give rise to neurons and oligodendrocytes. A pathological counterpart of this developmental function is seen in human glioma, wherein Olig2 is required for maintenance of stem-like cells that drive tumor growth. The mitogenic/gliomagenic functions of Olig2 are regulated by phosphorylation of a triple serine motif (S10, S13, and S14) in the amino terminus. Here, we identify a set of three serine/threonine protein kinases (glycogen synthase kinase 3α/ß [GSK3α/ß], casein kinase 2 [CK2], and cyclin-dependent kinases 1/2 [CDK1/2]) that are, collectively, both necessary and sufficient to phosphorylate the triple serine motif. We show that phosphorylation of the motif itself serves as a template to prime phosphorylation of additional serines and creates a highly charged "acid blob" in the amino terminus of Olig2. Finally, we show that small molecule inhibitors of this forward-feeding phosphorylation cascade have potential as glioma therapeutics.


Asunto(s)
Carcinogénesis/metabolismo , Carcinogénesis/patología , Glioma/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Animales , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Glioma/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Ratones , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína p53 Supresora de Tumor/metabolismo
16.
Nature ; 541(7638): 481-487, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28099414

RESUMEN

Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.


Asunto(s)
Astrocitos/clasificación , Astrocitos/patología , Muerte Celular , Sistema Nervioso Central/patología , Microglía/patología , Neuronas/patología , Animales , Astrocitos/metabolismo , Axotomía , Técnicas de Cultivo de Célula , Supervivencia Celular , Complemento C1q/metabolismo , Progresión de la Enfermedad , Humanos , Inflamación/patología , Interleucina-1alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Enfermedades Neurodegenerativas/patología , Oligodendroglía/patología , Fagocitosis , Fenotipo , Ratas , Ratas Sprague-Dawley , Sinapsis/patología , Toxinas Biológicas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Stem Cells ; 35(2): 311-315, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27882623

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is an X-linked disorder caused by mutation in the proteolipid protein-1 (PLP1) gene, which encodes the proteolipid protein of myelinating oligodendroglia. PMD exhibits phenotypic variability that reflects its considerable genotypic heterogeneity, but all forms of the disease result in central hypomyelination, associated in most cases with early neurological dysfunction, progressive deterioration, and ultimately death. PMD may present as a connatal, classic and transitional forms, or as the less severe spastic paraplegia type 2 and PLP-null phenotypes. These disorders are most often associated with duplications of the PLP1 gene, but can also be caused by coding and noncoding point mutations as well as full or partial deletion of the gene. A number of genetically-distinct but phenotypically-similar disorders of hypomyelination exist which, like PMD, lack any effective therapy. Yet as relatively pure CNS hypomyelinating disorders, with limited involvement of the PNS and relatively little attendant neuronal pathology, PMD and similar hypomyelinating disorders are attractive therapeutic targets for neural stem cell and glial progenitor cell transplantation, efforts at which are now underway in a number of research centers. Stem Cells 2017;35:311-315.


Asunto(s)
Enfermedad de Pelizaeus-Merzbacher/terapia , Trasplante de Células Madre , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Humanos , Mutación/genética , Vaina de Mielina/metabolismo , Enfermedad de Pelizaeus-Merzbacher/patología
18.
Cell Rep ; 16(11): 2838-2845, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27626655

RESUMEN

The basic helix-loop-helix (bHLH) transcription factor OLIG2 is a master regulator of oligodendroglial fate decisions and tumorigenic competence of glioma stem-like cells (GSCs). However, the molecular mechanisms underlying dysregulation of OLIG2 function during gliomagenesis remains poorly understood. Here, we show that OLIG2 modulates growth factor signaling in two distinct populations of GSCs, characterized by expression of either the epidermal growth factor receptor (EGFR) or platelet-derived growth factor receptor alpha (PDGFRα). Biochemical analyses of OLIG2 function in normal and malignant neural progenitors reveal a positive feedforward loop between OLIG2 and EGFR to sustain co-expression. Furthermore, loss of OLIG2 function results in mesenchymal transformation in PDGFRα(HIGH) GSCs, a phenomenon that appears to be circumscribed in EGFR(HIGH) GSCs. Exploitation of OLIG2's dual and antithetical, pro-mitotic (EGFR-driven), and lineage-specifying (PDGFRα-driven) functions by glioma cells appears to be critical for sustaining growth factor signaling and GSC molecular subtype.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/patología , Linaje de la Célula , Glioma/patología , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Animales , Neoplasias Encefálicas/metabolismo , Ciclo Celular , Núcleo Celular/metabolismo , Receptores ErbB/metabolismo , Glioma/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Factor de Transcripción 2 de los Oligodendrocitos , Fosforilación , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo
19.
Nat Commun ; 7: 11628, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27188978

RESUMEN

The pons controls crucial sensorimotor and autonomic functions. In humans, it grows sixfold postnatally and is a site of paediatric gliomas; however, the mechanisms of pontine growth remain poorly understood. We show that the murine pons quadruples in volume postnatally; growth is fastest during postnatal days 0-4 (P0-P4), preceding most myelination. We identify three postnatal proliferative compartments: ventricular, midline and parenchymal. We find no evidence of postnatal neurogenesis in the pons, but each progenitor compartment produces new astroglia and oligodendroglia; the latter expand 10- to 18-fold postnatally, and are derived mostly from the parenchyma. Nearly all parenchymal progenitors at P4 are Sox2(+)Olig2(+), but by P8 a Sox2(-) subpopulation emerges, suggesting a lineage progression from Sox2(+) 'early' to Sox2(-) 'late' oligodendrocyte progenitor. Fate mapping reveals that >90% of adult oligodendrocytes derive from P2-P3 Sox2(+) progenitors. These results demonstrate the importance of postnatal Sox2(+)Olig2(+) progenitors in pontine growth and oligodendrogenesis.


Asunto(s)
Células Precursoras de Oligodendrocitos/fisiología , Puente/crecimiento & desarrollo , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Proliferación Celular , Cuarto Ventrículo/citología , Ratones , Neurogénesis , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/fisiología , Puente/citología , Factores de Transcripción SOXB1/metabolismo
20.
Sci Transl Med ; 7(286): 286ra66, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25947161

RESUMEN

Astrocytes produce an assortment of signals that promote neuronal maturation according to a precise developmental timeline. Is this orchestrated timing and signaling altered in human neurodevelopmental disorders? To address this question, the astroglial lineage was investigated in two model systems of a developmental disorder with intellectual disability caused by mutant Harvey rat sarcoma viral oncogene homolog (HRAS) termed Costello syndrome: mutant HRAS human induced pluripotent stem cells (iPSCs) and transgenic mice. Human iPSCs derived from patients with Costello syndrome differentiated to astroglia more rapidly in vitro than those derived from wild-type cell lines with normal HRAS, exhibited hyperplasia, and also generated an abundance of extracellular matrix remodeling factors and proteoglycans. Acute treatment with a farnesyl transferase inhibitor and knockdown of the transcription factor SNAI2 reduced expression of several proteoglycans in Costello syndrome iPSC-derived astrocytes. Similarly, mice in which mutant HRAS was expressed selectively in astrocytes exhibited experience-independent increased accumulation of perineuronal net proteoglycans in cortex, as well as increased parvalbumin expression in interneurons, when compared to wild-type mice. Our data indicate that astrocytes expressing mutant HRAS dysregulate cortical maturation during development as shown by abnormal extracellular matrix remodeling and implicate excessive astrocyte-to-neuron signaling as a possible drug target for treating mental impairment and enhancing neuroplasticity.


Asunto(s)
Astrocitos/citología , Síndrome de Costello/metabolismo , Matriz Extracelular/metabolismo , Células Madre Pluripotentes Inducidas/citología , Transducción de Señal , Animales , Astrocitos/metabolismo , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Genes ras , Genotipo , Hipocampo/metabolismo , Humanos , Espectrometría de Masas , Ratones , Ratones Transgénicos , Mutación , Plasticidad Neuronal , Neuronas/citología , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Proteoglicanos/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA