Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3365, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664376

RESUMEN

Hedgehog (Hh) signaling relies on the primary cilium, a cell surface organelle that serves as a signaling hub for the cell. Using proximity labeling and quantitative proteomics, we identify Numb as a ciliary protein that positively regulates Hh signaling. Numb localizes to the ciliary pocket and acts as an endocytic adaptor to incorporate Ptch1 into clathrin-coated vesicles, thereby promoting Ptch1 exit from the cilium, a key step in Hh signaling activation. Numb loss impedes Sonic hedgehog (Shh)-induced Ptch1 exit from the cilium, resulting in reduced Hh signaling. Numb loss in spinal neural progenitors reduces Shh-induced differentiation into cell fates reliant on high Hh activity. Genetic ablation of Numb in the developing cerebellum impairs the proliferation of granule cell precursors, a Hh-dependent process, resulting in reduced cerebellar size. This study highlights Numb as a regulator of ciliary Ptch1 levels during Hh signal activation and demonstrates the key role of ciliary pocket-mediated endocytosis in cell signaling.


Asunto(s)
Cerebelo , Cilios , Proteínas Hedgehog , Proteínas del Tejido Nervioso , Receptor Patched-1 , Transducción de Señal , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Cilios/metabolismo , Animales , Receptor Patched-1/metabolismo , Receptor Patched-1/genética , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Cerebelo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Humanos , Endocitosis , Diferenciación Celular , Proliferación Celular , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Ratones Noqueados
2.
EMBO Rep ; 25(5): 2202-2219, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600346

RESUMEN

Neural progenitor cells within the cerebral cortex undergo a characteristic switch between symmetric self-renewing cell divisions early in development and asymmetric neurogenic divisions later. Yet, the mechanisms controlling this transition remain unclear. Previous work has shown that early but not late neural progenitor cells (NPCs) endogenously express the autism-linked transcription factor Foxp1, and both loss and gain of Foxp1 function can alter NPC activity and fate choices. Here, we show that premature loss of Foxp1 upregulates transcriptional programs regulating angiogenesis, glycolysis, and cellular responses to hypoxia. These changes coincide with a premature destabilization of HIF-1α, an elevation in HIF-1α target genes, including Vegfa in NPCs, and precocious vascular network development. In vitro experiments demonstrate that stabilization of HIF-1α in Foxp1-deficient NPCs rescues the premature differentiation phenotype and restores NPC maintenance. Our data indicate that the endogenous decline in Foxp1 expression activates the HIF-1α transcriptional program leading to changes in the tissue environment adjacent to NPCs, which, in turn, might alter their self-renewal and neurogenic capacities.


Asunto(s)
Corteza Cerebral , Factores de Transcripción Forkhead , Subunidad alfa del Factor 1 Inducible por Hipoxia , Células-Madre Neurales , Proteínas Represoras , Transducción de Señal , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Animales , Ratones , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Neovascularización Fisiológica/genética , Diferenciación Celular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Neurogénesis/genética , Glucólisis , Angiogénesis
3.
Trends Biochem Sci ; 48(3): 203-210, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36504139

RESUMEN

The process of evaluating and negotiating a tenure-track job offer is unstructured and highly variable, making it susceptible to bias and inequitable outcomes. We outline common aspects of and recommendations for negotiating an academic job offer in the life sciences to support equitable recruitment of diverse faculty.


Asunto(s)
Selección de Profesión , Empleo , Docentes , Negociación
4.
Development ; 148(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486668

RESUMEN

Birth defects result from interactions between genetic and environmental factors, but the mechanisms remain poorly understood. We find that mutations and teratogens interact in predictable ways to cause birth defects by changing target cell sensitivity to Hedgehog (Hh) ligands. These interactions converge on a membrane protein complex, the MMM complex, that promotes degradation of the Hh transducer Smoothened (SMO). Deficiency of the MMM component MOSMO results in elevated SMO and increased Hh signaling, causing multiple birth defects. In utero exposure to a teratogen that directly inhibits SMO reduces the penetrance and expressivity of birth defects in Mosmo-/- embryos. Additionally, tissues that develop normally in Mosmo-/- embryos are refractory to the teratogen. Thus, changes in the abundance of the protein target of a teratogen can change birth defect outcomes by quantitative shifts in Hh signaling. Consequently, small molecules that re-calibrate signaling strength could be harnessed to rescue structural birth defects.


Asunto(s)
Anomalías Inducidas por Medicamentos/genética , Interacción Gen-Ambiente , Proteínas Hedgehog/metabolismo , Penetrancia , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Transducción de Señal , Receptor Smoothened/genética , Receptor Smoothened/metabolismo
6.
Dev Cell ; 55(4): 432-449.e12, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32966817

RESUMEN

The etiology of congenital heart defects (CHDs), which are among the most common human birth defects, is poorly understood because of its complex genetic architecture. Here, we show that two genes implicated in CHDs, Megf8 and Mgrn1, interact genetically and biochemically to regulate the strength of Hedgehog signaling in target cells. MEGF8, a transmembrane protein, and MGRN1, a RING superfamily E3 ligase, assemble to form a receptor-like ubiquitin ligase complex that catalyzes the ubiquitination and degradation of the Hedgehog pathway transducer Smoothened. Homozygous Megf8 and Mgrn1 mutations increased Smoothened abundance and elevated sensitivity to Hedgehog ligands. While mice heterozygous for loss-of-function Megf8 or Mgrn1 mutations were normal, double heterozygous embryos exhibited an incompletely penetrant syndrome of CHDs with heterotaxy. Thus, genetic interactions can arise from biochemical mechanisms that calibrate morphogen signaling strength, a conclusion broadly relevant for the many human diseases in which oligogenic inheritance is emerging as a mechanism for heritability.


Asunto(s)
Corazón/embriología , Proteínas Hedgehog/metabolismo , Transducción de Señal , Ubiquitinación , Alelos , Animales , Embrión de Mamíferos/metabolismo , Epistasis Genética , Dosificación de Gen , Proteínas de la Membrana/metabolismo , Ratones , Mutación/genética , Células 3T3 NIH , Fenotipo , Unión Proteica , Receptor Smoothened/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
7.
Elife ; 82019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31657721

RESUMEN

Previously we proposed that transmission of the hedgehog signal across the plasma membrane by Smoothened is triggered by its interaction with cholesterol (Luchetti et al., 2016). But how is cholesterol, an abundant lipid, regulated tightly enough to control a signaling system that can cause birth defects and cancer? Using toxin-based sensors that distinguish between distinct pools of cholesterol, we find that Smoothened activation and Hedgehog signaling are driven by a biochemically-defined, small fraction of membrane cholesterol, termed accessible cholesterol. Increasing cholesterol accessibility by depletion of sphingomyelin, which sequesters cholesterol in complexes, amplifies Hedgehog signaling. Hedgehog ligands increase cholesterol accessibility in the membrane of the primary cilium by inactivating the transporter-like protein Patched 1. Trapping this accessible cholesterol blocks Hedgehog signal transmission across the membrane. Our work shows that the organization of cholesterol in the ciliary membrane can be modified by extracellular ligands to control the activity of cilia-localized signaling proteins.


Asunto(s)
Colesterol/metabolismo , Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos
8.
Development ; 146(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092502

RESUMEN

Signaling pathways that mediate cell-cell communication are essential for collective cell behaviors in multicellular systems. The hedgehog (HH) pathway, first discovered and elucidated in Drosophila, is one of these iconic signaling systems that plays many roles during embryogenesis and in adults; abnormal HH signaling can lead to birth defects and cancer. We review recent structural and biochemical studies that have advanced our understanding of the vertebrate HH pathway, focusing on the mechanisms by which the HH signal is received by patched on target cells, transduced across the cell membrane by smoothened, and transmitted to the nucleus by GLI proteins to influence gene-expression programs.


Asunto(s)
Proteínas Hedgehog/metabolismo , Vertebrados/metabolismo , Animales , Proteínas Hedgehog/genética , Humanos , Transducción de Señal/genética , Transducción de Señal/fisiología
9.
Sci Signal ; 11(516)2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29438014

RESUMEN

The morphogen Sonic Hedgehog (SHH) patterns tissues during development by directing cell fates in a concentration-dependent manner. The SHH signal is transmitted across the membrane of target cells by the heptahelical transmembrane protein Smoothened (SMO), which activates the GLI family of transcription factors through a mechanism that is undefined in vertebrates. Using CRISPR-edited null alleles and small-molecule inhibitors, we systematically analyzed the epistatic interactions between SMO and three proteins implicated in SMO signaling: the heterotrimeric G protein subunit GαS, the G protein-coupled receptor kinase 2 (GRK2), and the GαS-coupled receptor GPR161. Our experiments uncovered a signaling mechanism that modifies the sensitivity of target cells to SHH and consequently changes the shape of the SHH dose-response curve. In both fibroblasts and spinal neural progenitors, the loss of GPR161, previously implicated as an inhibitor of basal SHH signaling, increased the sensitivity of target cells across the entire spectrum of SHH concentrations. Even in cells lacking GPR161, GRK2 was required for SHH signaling, and Gαs, which promotes the activation of protein Kinase A (PKA), antagonized SHH signaling. We propose that the sensitivity of target cells to Hedgehog morphogens, and the consequent effects on gene expression and differentiation outcomes, can be controlled by signals from G protein-coupled receptors that converge on Gαs and PKA.


Asunto(s)
Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Receptor Smoothened/metabolismo , Animales , Células Cultivadas , Cromograninas/genética , Cromograninas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Epistasis Genética , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Ratones , Células 3T3 NIH , Receptores Acoplados a Proteínas G/genética , Receptor Smoothened/genética
11.
PLoS Biol ; 16(2): e2003127, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29389974

RESUMEN

During tissue development, multipotent progenitors differentiate into specific cell types in characteristic spatial and temporal patterns. We addressed the mechanism linking progenitor identity and differentiation rate in the neural tube, where motor neuron (MN) progenitors differentiate more rapidly than other progenitors. Using single cell transcriptomics, we defined the transcriptional changes associated with the transition of neural progenitors into MNs. Reconstruction of gene expression dynamics from these data indicate a pivotal role for the MN determinant Olig2 just prior to MN differentiation. Olig2 represses expression of the Notch signaling pathway effectors Hes1 and Hes5. Olig2 repression of Hes5 appears to be direct, via a conserved regulatory element within the Hes5 locus that restricts expression from MN progenitors. These findings reveal a tight coupling between the regulatory networks that control patterning and neuronal differentiation and demonstrate how Olig2 acts as the developmental pacemaker coordinating the spatial and temporal pattern of MN generation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Ciclo Celular/genética , Neuronas Motoras/citología , Neurogénesis/genética , Factor de Transcripción 2 de los Oligodendrocitos/fisiología , Proteínas Represoras/fisiología , Análisis de la Célula Individual , Factor de Transcripción HES-1/fisiología , Transcriptoma , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Colorantes Fluorescentes/metabolismo , Regulación de la Expresión Génica/fisiología , Genes Reporteros , Interneuronas/citología , Ratones Transgénicos , Factor de Transcripción 2 de los Oligodendrocitos/genética , Receptores Notch/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/genética , Transducción de Señal , Factor de Transcripción HES-1/genética
12.
Dev Cell ; 44(1): 113-129.e8, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29290584

RESUMEN

To uncover regulatory mechanisms in Hedgehog (Hh) signaling, we conducted genome-wide screens to identify positive and negative pathway components and validated top hits using multiple signaling and differentiation assays in two different cell types. Most positive regulators identified in our screens, including Rab34, Pdcl, and Tubd1, were involved in ciliary functions, confirming the central role for primary cilia in Hh signaling. Negative regulators identified included Megf8, Mgrn1, and an unannotated gene encoding a tetraspan protein we named Atthog. The function of these negative regulators converged on Smoothened (SMO), an oncoprotein that transduces the Hh signal across the membrane. In the absence of Atthog, SMO was stabilized at the cell surface and concentrated in the ciliary membrane, boosting cell sensitivity to the ligand Sonic Hedgehog (SHH) and consequently altering SHH-guided neural cell-fate decisions. Thus, we uncovered genes that modify the interpretation of morphogen signals by regulating protein-trafficking events in target cells.


Asunto(s)
Cilios/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteínas Hedgehog/farmacología , Neuronas/fisiología , Transducción de Señal/efectos de los fármacos , Receptor Smoothened/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Cilios/efectos de los fármacos , Fluorescencia , Proteínas de la Membrana/metabolismo , Ratones , Células 3T3 NIH , Neuronas/citología , Neuronas/efectos de los fármacos , Receptor Smoothened/genética
13.
Neuron ; 94(4): 790-799.e3, 2017 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-28434801

RESUMEN

Netrin1 has been proposed to act from the floor plate (FP) as a long-range diffusible chemoattractant for commissural axons in the embryonic spinal cord. However, netrin1 mRNA and protein are also present in neural progenitors within the ventricular zone (VZ), raising the question of which source of netrin1 promotes ventrally directed axon growth. Here, we use genetic approaches in mice to selectively remove netrin from different regions of the spinal cord. Our analyses show that the FP is not the source of netrin1 directing axons to the ventral midline, while local VZ-supplied netrin1 is required for this step. Furthermore, rather than being present in a gradient, netrin1 protein accumulates on the pial surface adjacent to the path of commissural axon extension. Thus, netrin1 does not act as a long-range secreted chemoattractant for commissural spinal axons but instead promotes ventrally directed axon outgrowth by haptotaxis, i.e., directed growth along an adhesive surface.


Asunto(s)
Orientación del Axón/genética , Axones/metabolismo , Factores de Crecimiento Nervioso/genética , Células-Madre Neurales/metabolismo , Médula Espinal/embriología , Proteínas Supresoras de Tumor/genética , Animales , Axones/ultraestructura , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Imagenología Tridimensional , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Microscopía Confocal , Factores de Crecimiento Nervioso/metabolismo , Netrina-1 , Neurogénesis/genética , ARN Mensajero/metabolismo , Médula Espinal/ultraestructura , Proteínas Supresoras de Tumor/metabolismo
14.
Elife ; 52016 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-27705744

RESUMEN

Cholesterol is necessary for the function of many G-protein coupled receptors (GPCRs). We find that cholesterol is not just necessary but also sufficient to activate signaling by the Hedgehog (Hh) pathway, a prominent cell-cell communication system in development. Cholesterol influences Hh signaling by directly activating Smoothened (SMO), an orphan GPCR that transmits the Hh signal across the membrane in all animals. Unlike many GPCRs, which are regulated by cholesterol through their heptahelical transmembrane domains, SMO is activated by cholesterol through its extracellular cysteine-rich domain (CRD). Residues shown to mediate cholesterol binding to the CRD in a recent structural analysis also dictate SMO activation, both in response to cholesterol and to native Hh ligands. Our results show that cholesterol can initiate signaling from the cell surface by engaging the extracellular domain of a GPCR and suggest that SMO activity may be regulated by local changes in cholesterol abundance or accessibility.


Asunto(s)
Colesterol/metabolismo , Erizos/metabolismo , Transducción de Señal , Receptor Smoothened/agonistas , Animales , Línea Celular , Células Epiteliales/fisiología , Fibroblastos/fisiología , Humanos , Ratones
15.
Dev Cell ; 33(4): 373-87, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25936505

RESUMEN

Throughout the developing nervous system, neural stem and progenitor cells give rise to diverse classes of neurons and glia in a spatially and temporally coordinated manner. In the ventral spinal cord, much of this diversity emerges through the morphogen actions of Sonic hedgehog (Shh). Interpretation of the Shh gradient depends on both the amount of ligand and duration of exposure, but the mechanisms permitting prolonged responses to Shh are not well understood. We demonstrate that Notch signaling plays an essential role in this process, enabling neural progenitors to attain sufficiently high levels of Shh pathway activity needed to direct the ventral-most cell fates. Notch activity regulates subcellular localization of the Shh receptor Patched1, gating the translocation of the key effector Smoothened to primary cilia and its downstream signaling activities. These data reveal an unexpected role for Notch shaping the interpretation of the Shh morphogen gradient and influencing cell fate determination.


Asunto(s)
Diferenciación Celular , Cilios/fisiología , Proteínas Hedgehog/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Receptores Notch/metabolismo , Células Madre/metabolismo , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Transgénicos , Neuroglía/citología , Neuroglía/metabolismo , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Receptor Smoothened , Médula Espinal/citología , Médula Espinal/metabolismo
16.
Cell Rep ; 6(1): 168-181, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24373970

RESUMEN

Gli proteins are transcriptional effectors of the Hedgehog (Hh) pathway in both normal development and cancer. We describe a program of multisite phosphorylation that regulates the conversion of Gli proteins into transcriptional activators. In the absence of Hh ligands, Gli activity is restrained by the direct phosphorylation of six conserved serine residues by protein kinase A (PKA), a master negative regulator of the Hh pathway. Activation of signaling leads to a global remodeling of the Gli phosphorylation landscape: the PKA target sites become dephosphorylated, while a second cluster of sites undergoes phosphorylation. The pattern of Gli phosphorylation can regulate Gli transcriptional activity in a graded fashion, suggesting a phosphorylation-based mechanism for how a gradient of Hh signaling in a morphogenetic field can be converted into a gradient of transcriptional activity.


Asunto(s)
Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Animales , Embrión de Pollo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/química , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Células 3T3 NIH , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Serina/metabolismo , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
17.
Dev Cell ; 25(5): 436-8, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23763945

RESUMEN

Voluntary motor control requires circuits in the brain to develop synchronously with spinal motor circuitry. In this issue of Developmental Cell,Reimer et al. (2013) demonstrate that this process is coordinated in zebrafish: dopamine released from descending projections modulates formation of motor neurons by attenuating the response of progenitors to Shh signaling.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Dopamina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas Motoras/citología , Regeneración , Animales
18.
Proc Natl Acad Sci U S A ; 109(20): 7882-7, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22543161

RESUMEN

Previous lineage analyses have shown that retinal progenitor cells (RPCs) are multipotent throughout development, and expression-profiling studies have shown a great deal of molecular heterogeneity among RPCs. To determine if the molecular heterogeneity predicts that an RPC will produce particular types of progeny, clonal lineage analysis was used to investigate the progeny of a subset of RPCs, those that express the basic helix-loop-helix transcription factor, Olig2. The embryonic Olig2(+) RPCs underwent terminal divisions, producing small clones with primarily two of the five cell types being made by the pool of RPCs at that time. The later, postnatal Olig2(+) RPCs also made terminal divisions, which were biased toward production of rod photoreceptors and amacrine cell interneurons. These data indicate that the multipotent progenitor pool is made up of distinctive types of RPCs, which have biases toward producing subsets of retinal neurons in a terminal division, with the types of neurons produced varying over time. This strategy is similar to that of the developing Drosophila melanogaster ventral nerve cord, with the Olig2(+) cells behaving as ganglion mother cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proteínas del Tejido Nervioso/metabolismo , Retina/citología , Células Madre/metabolismo , Animales , Electroporación , Inmunohistoquímica , Hibridación in Situ , Ratones , Factor de Transcripción 2 de los Oligodendrocitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...