Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(7): 1661-1676.e19, 2019 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-31835038

RESUMEN

Reliable detection of disseminated tumor cells and of the biodistribution of tumor-targeting therapeutic antibodies within the entire body has long been needed to better understand and treat cancer metastasis. Here, we developed an integrated pipeline for automated quantification of cancer metastases and therapeutic antibody targeting, named DeepMACT. First, we enhanced the fluorescent signal of cancer cells more than 100-fold by applying the vDISCO method to image metastasis in transparent mice. Second, we developed deep learning algorithms for automated quantification of metastases with an accuracy matching human expert manual annotation. Deep learning-based quantification in 5 different metastatic cancer models including breast, lung, and pancreatic cancer with distinct organotropisms allowed us to systematically analyze features such as size, shape, spatial distribution, and the degree to which metastases are targeted by a therapeutic monoclonal antibody in entire mice. DeepMACT can thus considerably improve the discovery of effective antibody-based therapeutics at the pre-clinical stage. VIDEO ABSTRACT.


Asunto(s)
Anticuerpos/uso terapéutico , Aprendizaje Profundo , Diagnóstico por Computador/métodos , Quimioterapia Asistida por Computador/métodos , Neoplasias/patología , Animales , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Programas Informáticos , Microambiente Tumoral
2.
Annu Rev Cell Dev Biol ; 35: 615-635, 2019 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-31590587

RESUMEN

Molecular cross talk between the nervous and vascular systems is necessary to maintain the correct coupling of organ structure and function. Molecular pathways shared by both systems are emerging as major players in the communication of the neuronal compartment with the endothelium. Here we review different aspects of this cross talk and how vessels influence the development and homeostasis of the nervous system. Beyond the classical role of the vasculature as a conduit to deliver oxygen and metabolites needed for the energy-demanding neuronal compartment, vessels emerge as powerful signaling systems that control and instruct a variety of cellular processes during the development of neurons and glia, such as migration, differentiation, and structural connectivity. Moreover, a broad spectrum of mild to severe vascular dysfunctions occur in various pathologies of the nervous system, suggesting that mild structural and functional changes at the neurovascular interface may underlie cognitive decline in many of these pathological conditions.


Asunto(s)
Sistema Nervioso Central/irrigación sanguínea , Neuroglía/citología , Neuronas/citología , Acoplamiento Neurovascular/fisiología , Sistema Nervioso Periférico/irrigación sanguínea , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Diferenciación Celular , Movimiento Celular , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Homeostasis/fisiología , Humanos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/embriología , Sistema Nervioso Periférico/metabolismo
3.
Cell ; 144(4): 566-76, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21335238

RESUMEN

TAp63α, a homolog of the p53 tumor suppressor, is a quality control factor in the female germline. Remarkably, already undamaged oocytes express high levels of the protein, suggesting that TAp63α's activity is under tight control of an inhibitory mechanism. Biochemical studies have proposed that inhibition requires the C-terminal transactivation inhibitory domain. However, the structural mechanism of TAp63α inhibition remains unknown. Here, we show that TAp63α is kept in an inactive dimeric state. We reveal that relief of inhibition leads to tetramer formation with ∼20-fold higher DNA affinity. In vivo, phosphorylation-triggered tetramerization of TAp63α is not reversible by dephosphorylation. Furthermore, we show that a helix in the oligomerization domain of p63 is crucial for tetramer stabilization and competes with the transactivation domain for the same binding site. Our results demonstrate how TAp63α is inhibited by complex domain-domain interactions that provide the basis for regulating quality control in oocytes.


Asunto(s)
Oocitos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Animales , ADN/metabolismo , Dimerización , Femenino , Rayos gamma , Ratones , Modelos Moleculares , Fosforilación , Multimerización de Proteína , Proteína p53 Supresora de Tumor/metabolismo
4.
EMBO Rep ; 22(2): e48961, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33512764

RESUMEN

Endothelial tip cells are essential for VEGF-induced angiogenesis, but underlying mechanisms are elusive. The Ena/VASP protein family, consisting of EVL, VASP, and Mena, plays a pivotal role in axon guidance. Given that axonal growth cones and endothelial tip cells share many common features, from the morphological to the molecular level, we investigated the role of Ena/VASP proteins in angiogenesis. EVL and VASP, but not Mena, are expressed in endothelial cells of the postnatal mouse retina. Global deletion of EVL (but not VASP) compromises the radial sprouting of the vascular plexus in mice. Similarly, endothelial-specific EVL deletion compromises the radial sprouting of the vascular plexus and reduces the endothelial tip cell density and filopodia formation. Gene sets involved in blood vessel development and angiogenesis are down-regulated in EVL-deficient P5-retinal endothelial cells. Consistently, EVL deletion impairs VEGF-induced endothelial cell proliferation and sprouting, and reduces the internalization and phosphorylation of VEGF receptor 2 and its downstream signaling via the MAPK/ERK pathway. Together, we show that endothelial EVL regulates sprouting angiogenesis via VEGF receptor-2 internalization and signaling.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Células Endoteliales , Neovascularización Fisiológica , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Animales , Células Endoteliales/metabolismo , Ratones , Morfogénesis , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Am J Physiol Cell Physiol ; 318(4): C719-C731, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31967857

RESUMEN

Carotid body (CB) type I cells sense the blood Po2 and generate a nervous signal for stimulating ventilation and circulation when blood oxygen levels decline. Three oxygen-sensing enzyme complexes may be used for this purpose: 1) mitochondrial electron transport chain metabolism, 2) heme oxygenase 2 (HO-2)-generating CO, and/or 3) an NAD(P)H oxidase (NOX). We hypothesize that intracellular redox changes are the link between the sensor and nervous signals. To test this hypothesis type I cell autofluorescence of flavoproteins (Fp) and NAD(P)H within the mouse CB ex vivo was recorded as Fp/(Fp+NAD(P)H) redox ratio. CB type I cell redox ratio transiently declined with the onset of hypoxia. Upon reoxygenation, CB type I cells showed a significantly increased redox ratio. As a control organ, the non-oxygen-sensing sympathetic superior cervical ganglion (SCG) showed a continuously reduced redox ratio upon hypoxia. CN-, diphenyleneiodonium, or reactive oxygen species influenced chemoreceptor discharge (CND) with subsequent loss of O2 sensitivity and inhibited hypoxic Fp reduction only in the CB but not in SCG Fp, indicating a specific role of Fp in the oxygen-sensing process. Hypoxia-induced changes in CB type I cell redox ratio affected peptidyl prolyl isomerase Pin1, which is believed to colocalize with the NADPH oxidase subunit p47phox in the cell membrane to trigger the opening of potassium channels. We postulate that hypoxia-induced changes in the Fp-mediated redox ratio of the CB regulate the Pin1/p47phox tandem to alter type I cell potassium channels and therewith CND.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Quimiorreceptoras/metabolismo , Flavoproteínas/metabolismo , Hipoxia/metabolismo , Pulmón/metabolismo , Ratones , Mitocondrias/metabolismo , Canales de Potasio/metabolismo
6.
Cell Mol Life Sci ; 76(11): 2133-2169, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30937469

RESUMEN

To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.


Asunto(s)
Proteínas Portadoras/metabolismo , Red Nerviosa/fisiología , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Proteínas Nucleares/metabolismo , Receptores AMPA/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas del Huevo/genética , Proteínas del Huevo/metabolismo , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Redes Neurales de la Computación , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Transporte de Proteínas , Receptores AMPA/genética , Sinapsis/metabolismo , Transmisión Sináptica
7.
Biol Chem ; 399(5): 453-465, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29337689

RESUMEN

Employing hippocampal synaptosomes from amyloid precursor protein (APP)-deleted mice we analyzed the immediate effects of amyloid beta peptide 42 (Aß42) peptide in its oligomeric or fibrillar assembly or of soluble amyloid precursor protein alpha (sAPPα) protein on their bioenergetic activity. Upon administration of oligomeric Aß42 peptide for 30 min we observed a robust decrease both in mitochondrial activity and in mitochondrial membrane potential (MMP). In contrast the respective fibrillary or scrambled peptides showed no effect, indicating that inhibition strictly depends on the oligomerization status of the peptide. Hippocampal synaptosomes from old APP-KO mice revealed a further reduction of their already impaired bioenergetic activity upon incubation with 10 µm Aß42 peptide. In addition we evaluated the influence of the sAPPα protein on mitochondrial activity of hippocampal synaptosomes derived from young or old APP-KO animals. In neither case 20 nm nor 200 nm sAPPα protein had an effect on mitochondrial metabolic activity. Our findings demonstrate that hippocampal synaptosomes derived from APP-KO mice are a most suitable model system to evaluate the impact of Aß42 peptide on its bioenergetic activity and to further elucidate the molecular mechanisms underlying the impairments by oligomeric Aß42 on mitochondrial function. Our data demonstrate that extracellular Aß42 peptide is taken up into synaptosomes where it immediately attenuates mitochondrial activity.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Sinaptosomas/metabolismo , Precursor de Proteína beta-Amiloide/deficiencia , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo
8.
Development ; 141(3): 661-73, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24449842

RESUMEN

The proper functioning of the dopaminergic system requires the coordinated formation of projections extending from dopaminergic neurons in the substantia nigra (SN), ventral tegmental area (VTA) and retrorubral field to a wide array of forebrain targets including the striatum, nucleus accumbens and prefrontal cortex. The mechanisms controlling the assembly of these distinct dopaminergic cell clusters are not well understood. Here, we have investigated in detail the migratory behavior of dopaminergic neurons giving rise to either the SN or the medial VTA using genetic inducible fate mapping, ultramicroscopy, time-lapse imaging, slice culture and analysis of mouse mutants. We demonstrate that neurons destined for the SN migrate first radially and then tangentially, whereas neurons destined for the medial VTA undergo primarily radial migration. We show that tangentially migrating dopaminergic neurons express the components of the reelin signaling pathway, whereas dopaminergic neurons in their initial, radial migration phase express CXC chemokine receptor 4 (CXCR4), the receptor for the chemokine CXC motif ligand 12 (CXCL12). Perturbation of reelin signaling interferes with the speed and orientation of tangentially, but not radially, migrating dopaminergic neurons and results in severe defects in the formation of the SN. By contrast, CXCR4/CXCL12 signaling modulates the initial migration of dopaminergic neurons. With this study, we provide the first molecular and functional characterization of the distinct migratory pathways taken by dopaminergic neurons destined for SN and VTA, and uncover mechanisms that regulate different migratory behaviors of dopaminergic neurons.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Quimiocina CXCL12/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Linaje de la Célula , Desarrollo Embrionario , Ligandos , Ratones , Ratones Noqueados , Modelos Biológicos , Receptores CXCR4/metabolismo , Proteína Reelina , Transducción de Señal , Sustancia Negra/citología , Imagen de Lapso de Tiempo , Área Tegmental Ventral/citología
9.
PLoS Comput Biol ; 12(4): e1004832, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27092780

RESUMEN

The hallmarks of Alzheimer's disease (AD) are characterized by cognitive decline and behavioral changes. The most prominent brain region affected by the progression of AD is the hippocampal formation. The pathogenesis involves a successive loss of hippocampal neurons accompanied by a decline in learning and memory consolidation mainly attributed to an accumulation of senile plaques. The amyloid precursor protein (APP) has been identified as precursor of Aß-peptides, the main constituents of senile plaques. Until now, little is known about the physiological function of APP within the central nervous system. The allocation of APP to the proteome of the highly dynamic presynaptic active zone (PAZ) highlights APP as a yet unknown player in neuronal communication and signaling. In this study, we analyze the impact of APP deletion on the hippocampal PAZ proteome. The native hippocampal PAZ derived from APP mouse mutants (APP-KOs and NexCreAPP/APLP2-cDKOs) was isolated by subcellular fractionation and immunopurification. Subsequently, an isobaric labeling was performed using TMT6 for protein identification and quantification by high-resolution mass spectrometry. We combine bioinformatics tools and biochemical approaches to address the proteomics dataset and to understand the role of individual proteins. The impact of APP deletion on the hippocampal PAZ proteome was visualized by creating protein-protein interaction (PPI) networks that incorporated APP into the synaptic vesicle cycle, cytoskeletal organization, and calcium-homeostasis. The combination of subcellular fractionation, immunopurification, proteomic analysis, and bioinformatics allowed us to identify APP as structural and functional regulator in a context-sensitive manner within the hippocampal active zone network.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Biología Computacional , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terminales Presinápticos/metabolismo , Mapas de Interacción de Proteínas , Proteoma/metabolismo , Sinapsis/metabolismo
10.
Nature ; 472(7343): 356-60, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21460838

RESUMEN

Coordinated migration of neurons in the developing and adult brain is essential for its proper function. The secreted glycoprotein Reelin (also known as RELN) guides migration of neurons by binding to two lipoprotein receptors, the very-low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2, also known as LRP8). Loss of Reelin function in humans results in the severe developmental disorder lissencephaly and it has also been associated with other neurological disorders such as epilepsy, schizophrenia and Alzheimer's disease. The molecular mechanisms by which Reelin activates its receptors and controls cellular functions are largely unknown. Here we show that the neuronal guidance cues ephrin B proteins are essential for Reelin signalling during the development of laminated structures in the brain. We show that ephrin Bs genetically interact with Reelin. Notably, compound mouse mutants (Reln(+/-); Efnb3(-/-) or Reln(+/-); Efnb2(-/-)) and triple ephrin B1, B2, B3 knockouts show neuronal migration defects that recapitulate the ones observed in the neocortex, hippocampus and cerebellum of the reeler mouse. Mechanistically, we show that Reelin binds to the extracellular domain of ephrin Bs, which associate at the membrane with VLDLR and ApoER2 in neurons. Clustering of ephrin Bs leads to the recruitment and phosphorylation of Dab1 which is necessary for Reelin signalling. Conversely, loss of function of ephrin Bs severely impairs Reelin-induced Dab1 phosphorylation. Importantly, activation of ephrin Bs can rescue the reeler neuronal migration defects in the absence of Reelin protein. Together, our results identify ephrin Bs as essential components of the Reelin receptor/signalling pathway to control neuronal migration during the development of the nervous system.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Corteza Cerebral/citología , Efrinas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Animales , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Efrina-B1/deficiencia , Efrina-B1/genética , Efrina-B1/metabolismo , Efrina-B2/deficiencia , Efrina-B2/genética , Efrina-B2/metabolismo , Efrina-B3/deficiencia , Efrina-B3/genética , Efrina-B3/metabolismo , Efrinas/deficiencia , Efrinas/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ligandos , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Fenotipo , Fosforilación , Unión Proteica , Receptores de LDL/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética
11.
Proc Natl Acad Sci U S A ; 111(6): 2194-9, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24469813

RESUMEN

Mammalian plexins constitute a family of transmembrane receptors for semaphorins and represent critical regulators of various processes during development of the nervous, cardiovascular, skeletal, and renal system. In vitro studies have shown that plexins exert their effects via an intracellular R-Ras/M-Ras GTPase-activating protein (GAP) domain or by activation of RhoA through interaction with Rho guanine nucleotide exchange factor proteins. However, which of these signaling pathways are relevant for plexin functions in vivo is largely unknown. Using an allelic series of transgenic mice, we show that the GAP domain of plexins constitutes their key signaling module during development. Mice in which endogenous Plexin-B2 or Plexin-D1 is replaced by transgenic versions harboring mutations in the GAP domain recapitulate the phenotypes of the respective null mutants in the developing nervous, vascular, and skeletal system. We further provide genetic evidence that, unexpectedly, the GAP domain-mediated developmental functions of plexins are not brought about via R-Ras and M-Ras inactivation. In contrast to the GAP domain mutants, Plexin-B2 transgenic mice defective in Rho guanine nucleotide exchange factor binding are viable and fertile but exhibit abnormal development of the liver vasculature. Our genetic analyses uncover the in vivo context-dependence and functional specificity of individual plexin-mediated signaling pathways during development.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal/genética , Animales , Ratones , Ratones Transgénicos
12.
Acta Neuropathol ; 131(2): 281-298, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26687980

RESUMEN

Remyelination in multiple sclerosis (MS) lesions often remains incomplete despite the presence of oligodendrocyte progenitor cells (OPCs). Amongst other factors, successful remyelination depends on the phagocytic clearance of myelin debris. However, the proteins in myelin debris that act as potent and selective inhibitors on OPC differentiation and inhibit CNS remyelination remain unknown. Here, we identify the transmembrane signalling protein EphrinB3 as important mediator of this inhibition, using a protein analytical approach in combination with a primary rodent OPC assay. In the presence of EphrinB3, OPCs fail to differentiate. In a rat model of remyelination, infusion of EphrinB3 inhibits remyelination. In contrast, masking EphrinB3 epitopes using antibodies promotes remyelination. Finally, we identify EphrinB3 in MS lesions and demonstrate that MS lesion extracts inhibit OPC differentiation while antibody-mediated masking of EphrinB3 epitopes promotes it. Our findings suggest that EphrinB3 could be a target for therapies aiming at promoting remyelination in demyelinating disease.


Asunto(s)
Efrina-B3/metabolismo , Esclerosis Múltiple/metabolismo , Vaina de Mielina/metabolismo , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Efrina-B3/genética , Epítopos/metabolismo , Femenino , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Noqueados , Esclerosis Múltiple/patología , Vaina de Mielina/patología , Regeneración Nerviosa/fisiología , Células-Madre Neurales/patología , Neurogénesis/fisiología , Oligodendroglía/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptor EphA4/metabolismo
13.
Stem Cells ; 33(1): 253-64, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25205248

RESUMEN

Nerve cells are continuously generated from stem cells in the adult mammalian subventricular zone (SVZ) and hippocampal dentate gyrus. We have previously noted that stem/progenitor cells in the SVZ and the subgranular layer (SGL) of the dentate gyrus express high levels of plasma membrane-bound nucleoside triphosphate diphosphohydrolase 2 (NTPDase2), an ectoenzyme that hydrolyzes extracellular nucleoside diphosphates and triphosphates. We inferred that deletion of NTPDase2 would increase local extracellular nucleoside triphosphate concentrations perturbing purinergic signaling and boosting progenitor cell proliferation and neurogenesis. Using newly generated mice globally null for Entpd2, we demonstrate that NTPDase2 is the major ectonucleotidase in these progenitor cell-rich areas. Using BrdU-labeling protocols, we have measured stem cell proliferation and determined long-term survival of cell progeny under basal conditions. Brains of Entpd2 null mice revealed increased progenitor cell proliferation in both the SVZ and the SGL. However, this occurred without noteworthy alterations in long-term progeny survival. The hippocampal stem cell pool and the pool of the intermediate progenitor type-2 cells clearly expanded. However, substantive proportions of these proliferating cells were lost during expansion at around type-3 stage. Cell loss was paralleled by decreases in cAMP response element-binding protein phosphorylation in the doublecortin-positive progenitor cell population and by an increase in labeling for activated caspase-3 levels. We propose that NTPDase2 has functionality in scavenging mitogenic extracellular nucleoside triphosphates in neurogenic niches of the adult brain, thereby acting as a homeostatic regulator of nucleotide-mediated neural progenitor cell proliferation and expansion.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Encéfalo/citología , Células-Madre Neurales/citología , Nicho de Células Madre/fisiología , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Proliferación Celular/fisiología , Inmunohistoquímica , Ratones , Células-Madre Neurales/enzimología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Transducción de Señal
14.
Nature ; 465(7297): 487-91, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20445540

RESUMEN

The formation and guidance of specialized endothelial tip cells is essential for both developmental and pathological angiogenesis. Notch-1 signalling regulates the generation of tip cells, which respond to gradients of vascular endothelial growth factor (VEGF-A). The molecular cues and signalling pathways that control the guidance of tip cells are poorly understood. Bidirectional signalling by Eph receptors and ephrin ligands represents one of the most important guidance cues involved in axon path finding. Here we show that ephrin-B2 reverse signalling involving PDZ interactions regulates endothelial tip cell guidance to control angiogenic sprouting and branching in physiological and pathological angiogenesis. In vivo, ephrin-B2 PDZ-signalling-deficient mice (ephrin-B2DeltaV) exhibit a reduced number of tip cells with fewer filopodial extensions at the vascular front in the mouse retina. In pathological settings, impaired PDZ signalling decreases tumour vascularization and growth. Mechanistically, we show that ephrin-B2 controls VEGF receptor (VEGFR)-2 internalization and signalling. Importantly, internalization of VEGFR2 is necessary for activation and downstream signalling of the receptor and is required for VEGF-induced tip cell filopodial extension. Together, our results suggest that ephrin-B2 at the tip cell filopodia regulates the proper spatial activation of VEGFR2 endocytosis and signalling to direct filopodial extension. Blocking ephrin-B2 reverse signalling may be an attractive alternative or combinatorial anti-angiogenic therapy strategy to disrupt VEGFR2 function in tumour angiogenesis.


Asunto(s)
Astrocitoma/irrigación sanguínea , Astrocitoma/metabolismo , Efrina-B2/metabolismo , Neovascularización Patológica , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Astrocitoma/patología , Encéfalo/irrigación sanguínea , Células Cultivadas , Endocitosis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Efrina-B2/deficiencia , Efrina-B2/genética , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neovascularización Fisiológica , Seudópodos/metabolismo , Retina , Vasos Retinianos/citología , Vasos Retinianos/fisiología , Transducción de Señal
15.
Purinergic Signal ; 11(1): 155-60, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25504514

RESUMEN

Eye formation in vertebrates is controlled by a conserved pattern of molecular networks. Homeobox transcription factors are crucially involved in the establishment and maintenance of the retina. A previous study of Massé et al. (Nature, 449: 1058-62, 2007) using morpholino knockdown identified the ectonucleotidase NTPDase2 and the P2Y1 receptor as essential elements for eye formation in embryos of the clawed frog Xenopus laevis. In order to investigate whether a similarly essential mechanism would be active in mammalian eye development, we analyzed mice KO for Entpd2 or P2ry1 as well as double KO for Entpd2/P2ry1. These mice developed normal eyes. In order to identify potential deficits in the molecular identity or in the arrangement of the cellular elements of the retina, we performed an immunohistological analysis using a variety of retinal markers. The analysis of single and double KO mice demonstrated that NTPDase2 and P2Y1 receptors are not required for murine eye formation, as previously shown for eye development in Xenopus laevis.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Ojo/embriología , Organogénesis/genética , Receptores Purinérgicos P2Y1/metabolismo , Retina/embriología , Adenosina Trifosfatasas/genética , Animales , Ojo/metabolismo , Ratones , Ratones Noqueados , Receptores Purinérgicos P2Y1/genética , Retina/metabolismo
16.
Adv Exp Med Biol ; 860: 55-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26303467

RESUMEN

Reactive oxygen species (ROS) generated by the NADPH oxidase have been proposed to play an important role in the carotid body (CB) oxygen sensing process (Cross et al. 1990). Up to now it remains unclear whether hypoxia causes an increase or decrease of CB ROS levels. We transfected CBs with the ROS sensitive HSP-FRET construct and subsequently measured the intracellular redox state by means of Förster resonance energy transfer (FRET) microscopy. In a previous study we found both increasing and decreasing ROS levels under hypoxic conditions. The transition from decreasing to increasing ROS levels coincided with the change of the caging system from ambient environment caging (AEC) to individually ventilated caging (IVC) (Bernardini A, Brockmeier U, Metzen E, Berchner-Pfannschmidt U, Harde E, Acker-Palmer A, Papkovsky D, Acker H, Fandrey J, Type I cell ROS kinetics under hypoxia in the intact mouse carotid body ex vivo: a FRET based study. Am J Physiol Cell Physiol. doi: 10.1152/ajpcell.00370.2013 , 2014). In this work we analyze hypoxia induced ROS reaction of animals from an IVC system that had been exposed to AEC conditions for 5 days. The results further support the hypothesis of an important impact of the caging system on CB ROS reaction.


Asunto(s)
Cuerpo Carotídeo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Transferencia Resonante de Energía de Fluorescencia , Potenciales de la Membrana
17.
EMBO J ; 29(16): 2677-88, 2010 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-20717138

RESUMEN

The spatial organization of transmembrane receptors is a critical step in signal transduction and receptor trafficking in cells. Transmembrane receptors engage in lateral homotypic and heterotypic cis-interactions as well as intercellular trans-interactions that result in the formation of signalling foci for the initiation of different signalling networks. Several aspects of ligand-induced receptor clustering and association with signalling proteins are also influenced by the lipid composition of membranes. Thus, lipid microdomains have a function in tuning the activity of many transmembrane receptors by positively or negatively affecting receptor clustering and signal transduction. We review the current knowledge about the functions of clustering of transmembrane receptors and lipid-protein interactions important for the spatial organization of signalling at the membrane.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Humanos , Microdominios de Membrana/metabolismo
18.
Neuron ; 112(2): 175-177, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38237553

RESUMEN

In this issue of Neuron, Bhat et al.1 unveil the temporary reawakening of an embryonic guidance program, which facilitates the alignment of blood neovessels, creating a supportive "bridge" microenvironment for axon regrowth and tissue regeneration after peripheral nervous system (PNS) injury.


Asunto(s)
Axones , Regeneración Nerviosa , Axones/fisiología , Regeneración Nerviosa/fisiología , Neuronas , Sistema Nervioso Periférico/fisiología
19.
J Biol Chem ; 287(25): 21224-32, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22539351

RESUMEN

The brain-specific isoform carnitine palmitoyltransferase 1C (CPT1C) has been implicated in the hypothalamic regulation of food intake and energy homeostasis. Nevertheless, its molecular function is not completely understood, and its role in other brain areas is unknown. We demonstrate that CPT1C is expressed in pyramidal neurons of the hippocampus and is located in the endoplasmic reticulum throughout the neuron, even inside dendritic spines. We used molecular, cellular, and behavioral approaches to determine CPT1C function. First, we analyzed the implication of CPT1C in ceramide metabolism. CPT1C overexpression in primary hippocampal cultured neurons increased ceramide levels, whereas in CPT1C-deficient neurons, ceramide levels were diminished. Correspondingly, CPT1C knock-out (KO) mice showed reduced ceramide levels in the hippocampus. At the cellular level, CPT1C deficiency altered dendritic spine morphology by increasing immature filopodia and reducing mature mushroom and stubby spines. Total protrusion density and spine head area in mature spines were unaffected. Treatment of cultured neurons with exogenous ceramide reverted the KO phenotype, as did ectopic overexpression of CPT1C, indicating that CPT1C regulation of spine maturation is mediated by ceramide. To study the repercussions of the KO phenotype on cognition, we performed the hippocampus-dependent Morris water maze test on mice. Results show that CPT1C deficiency strongly impairs spatial learning. All of these results demonstrate that CPT1C regulates the levels of ceramide in the endoplasmic reticulum of hippocampal neurons, and this is a relevant mechanism for the correct maturation of dendritic spines and for proper spatial learning.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Ceramidas/metabolismo , Dendritas/enzimología , Metabolismo Energético/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Metabolismo de los Lípidos/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Células Piramidales/enzimología , Animales , Conducta Animal/fisiología , Carnitina O-Palmitoiltransferasa/genética , Células Cultivadas , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/genética , Errores Innatos del Metabolismo Lipídico/enzimología , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/patología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Células Piramidales/citología
20.
Sci Rep ; 13(1): 20497, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993550

RESUMEN

Dendritic spines are considered a morphological proxy for excitatory synapses, rendering them a target of many different lines of research. Over recent years, it has become possible to simultaneously image large numbers of dendritic spines in 3D volumes of neural tissue. In contrast, currently no automated method for 3D spine detection exists that comes close to the detection performance reached by human experts. However, exploiting such datasets requires new tools for the fully automated detection and analysis of large numbers of spines. Here, we developed an efficient analysis pipeline to detect large numbers of dendritic spines in volumetric fluorescence imaging data acquired by two-photon imaging in vivo. The core of our pipeline is a deep convolutional neural network that was pretrained on a general-purpose image library and then optimized on the spine detection task. This transfer learning approach is data efficient while achieving a high detection precision. To train and validate the model we generated a labeled dataset using five human expert annotators to account for the variability in human spine detection. The pipeline enables fully automated dendritic spine detection reaching a performance slightly below that of the human experts. Our method for spine detection is fast, accurate and robust, and thus well suited for large-scale datasets with thousands of spines. The code is easily applicable to new datasets, achieving high detection performance, even without any retraining or adjustment of model parameters.


Asunto(s)
Espinas Dendríticas , Tejido Nervioso , Humanos , Redes Neurales de la Computación , Imagenología Tridimensional/métodos , Microscopía Confocal/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA