Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(17): e108780, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35815410

RESUMEN

Schwann cell precursors (SCPs) are nerve-associated progenitors that can generate myelinating and non-myelinating Schwann cells but also are multipotent like the neural crest cells from which they originate. SCPs are omnipresent along outgrowing peripheral nerves throughout the body of vertebrate embryos. By using single-cell transcriptomics to generate a gene expression atlas of the entire neural crest lineage, we show that early SCPs and late migratory crest cells have similar transcriptional profiles characterised by a multipotent "hub" state containing cells biased towards traditional neural crest fates. SCPs keep diverging from the neural crest after being primed towards terminal Schwann cells and other fates, with different subtypes residing in distinct anatomical locations. Functional experiments using CRISPR-Cas9 loss-of-function further show that knockout of the common "hub" gene Sox8 causes defects in neural crest-derived cells along peripheral nerves by facilitating differentiation of SCPs towards sympathoadrenal fates. Finally, specific tumour populations found in melanoma, neurofibroma and neuroblastoma map to different stages of SCP/Schwann cell development. Overall, SCPs resemble migrating neural crest cells that maintain multipotency and become transcriptionally primed towards distinct lineages.


Asunto(s)
Cresta Neural , Células de Schwann , Diferenciación Celular/fisiología , Neurogénesis/fisiología , Nervios Periféricos , Células de Schwann/metabolismo
2.
Am J Hum Genet ; 109(9): 1713-1723, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35948005

RESUMEN

The leucine-rich glioma-inactivated (LGI) family consists of four highly conserved paralogous genes, LGI1-4, that are highly expressed in mammalian central and/or peripheral nervous systems. LGI1 antibodies are detected in subjects with autoimmune limbic encephalitis and peripheral nerve hyperexcitability syndromes (PNHSs) such as Isaacs and Morvan syndromes. Pathogenic variations of LGI1 and LGI4 are associated with neurological disorders as disease traits including familial temporal lobe epilepsy and neurogenic arthrogryposis multiplex congenita 1 with myelin defects, respectively. No human disease has been reported associated with either LGI2 or LGI3. We implemented exome sequencing and family-based genomics to identify individuals with deleterious variants in LGI3 and utilized GeneMatcher to connect practitioners and researchers worldwide to investigate the clinical and electrophysiological phenotype in affected subjects. We also generated Lgi3-null mice and performed peripheral nerve dissection and immunohistochemistry to examine the juxtaparanode LGI3 microarchitecture. As a result, we identified 16 individuals from eight unrelated families with loss-of-function (LoF) bi-allelic variants in LGI3. Deep phenotypic characterization showed LGI3 LoF causes a potentially clinically recognizable PNHS trait characterized by global developmental delay, intellectual disability, distal deformities with diminished reflexes, visible facial myokymia, and distinctive electromyographic features suggestive of motor nerve instability. Lgi3-null mice showed reduced and mis-localized Kv1 channel complexes in myelinated peripheral axons. Our data demonstrate bi-allelic LoF variants in LGI3 cause a clinically distinguishable disease trait of PNHS, most likely caused by disturbed Kv1 channel distribution in the absence of LGI3.


Asunto(s)
Miocimia , Proteínas del Tejido Nervioso , Animales , Autoanticuerpos , Axones , Genómica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mamíferos/genética , Ratones , Proteínas del Tejido Nervioso/genética , Fenotipo , Genética Inversa
3.
Genes Dev ; 28(3): 290-303, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24493648

RESUMEN

Myelination depends on the synthesis of large amounts of myelin transcripts and proteins and is controlled by Nrg1/ErbB/Shp2 signaling. We developed a novel pulse labeling strategy based on stable isotope labeling with amino acids in cell culture (SILAC) to measure the dynamics of myelin protein production in mice. We found that protein synthesis is dampened in the maturing postnatal peripheral nervous system, and myelination then slows down. Remarkably, sustained activation of MAPK signaling by expression of the Mek1DD allele in mice overcomes the signals that end myelination, resulting in continuous myelin growth. MAPK activation leads to minor changes in transcript levels but massively up-regulates protein production. Pharmacological interference in vivo demonstrates that the effects of activated MAPK signaling on translation are mediated by mTOR-independent mechanisms but in part also by mTOR-dependent mechanisms. Previous work demonstrated that loss of ErbB3/Shp2 signaling impairs Schwann cell development and disrupts the myelination program. We found that activated MAPK signaling strikingly compensates for the absence of ErbB3 or Shp2 during Schwann cell development and myelination.


Asunto(s)
Diferenciación Celular , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Vaina de Mielina/metabolismo , Neurregulina-1/metabolismo , Receptor ErbB-3/metabolismo , Células de Schwann/citología , Alelos , Animales , Regulación de la Expresión Génica/genética , MAP Quinasa Quinasa 1/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Microscopía Electrónica de Transmisión , Complejos Multiproteicos , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptor ErbB-3/genética , Células de Schwann/ultraestructura , Transducción de Señal , Serina-Treonina Quinasas TOR
4.
Glia ; 69(11): 2605-2617, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34288120

RESUMEN

Disruption of axon-glia interactions in the peripheral nervous system has emerged as a major cause of arthrogryposis multiplex congenita (AMC), a condition characterized by multiple congenital postural abnormalities involving the major joints. Several genes crucially important to the biology of Schwann cells have now been implicated with AMC. One such gene is LGI4 which encodes a secreted glycoprotein. LGI4 is expressed and secreted by Schwann cells and binds its receptor ADAM22 on the axonal membrane to drive myelination. Homozygous mutations in LGI4 or ADAM22 results in severe congenital hypomyelination and joint contractures in mice. Recently bi-allelic LGI4 loss of function mutations has been described in three unrelated families with severe AMC. Two individuals in a fourth, non-consanguineous family were found to be compound heterozygous for two LGI4 missense mutations. It is not known how these missense mutations affect the biology of LGI4. Here we investigated whether these missense mutations affected the secretion of the protein, its ADAM22 binding capacity, or its myelination-promoting function. We demonstrate that the mutations largely affect the progression of the mutant protein through the endomembrane system resulting in severely reduced expression. Importantly, binding to ADAM22 and myelination-promoting activity appear largely unaffected, suggesting that treatment with chemical chaperones to improve secretion of the mutant proteins might prove beneficial.


Asunto(s)
Artrogriposis , Animales , Artrogriposis/genética , Artrogriposis/metabolismo , Axones/metabolismo , Humanos , Ratones , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células de Schwann/metabolismo
5.
J Neurosci ; 37(16): 4255-4269, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28320842

RESUMEN

After nerve injury, Schwann cells convert to a phenotype specialized to promote repair. But during the slow process of axonal regrowth, these repair Schwann cells gradually lose their regeneration-supportive features and eventually die. Although this is a key reason for the frequent regeneration failures in humans, the transcriptional mechanisms that control long-term survival and phenotype of repair cells have not been studied, and the molecular signaling underlying their decline is obscure. We show, in mice, that Schwann cell STAT3 has a dual role. It supports the long-term survival of repair Schwann cells and is required for the maintenance of repair Schwann cell properties. In contrast, STAT3 is less important for the initial generation of repair Schwann cells after injury. In repair Schwann cells, we find that Schwann cell STAT3 activation by Tyr705 phosphorylation is sustained during long-term denervation. STAT3 is required for maintaining autocrine Schwann cell survival signaling, and inactivation of Schwann cell STAT3 results in a striking loss of repair cells from chronically denervated distal stumps. STAT3 inactivation also results in abnormal morphology of repair cells and regeneration tracks, and failure to sustain expression of repair cell markers, including Shh, GDNF, and BDNF. Because Schwann cell development proceeds normally without STAT3, the function of this factor appears restricted to Schwann cells after injury. This identification of transcriptional mechanisms that support long-term survival and differentiation of repair cells will help identify, and eventually correct, the failures that lead to the deterioration of this important cell population.SIGNIFICANCE STATEMENT Although injured peripheral nerves contain repair Schwann cells that provide signals and spatial clues for promoting regeneration, the clinical outcome after nerve damage is frequently poor. A key reason for this is that, during the slow growth of axons through the proximal parts of injured nerves repair, Schwann cells gradually lose regeneration-supporting features and eventually die. Identification of signals that sustain repair cells is therefore an important goal. We have found that in mice the transcription factor STAT3 protects these cells from death and contributes to maintaining the molecular and morphological repair phenotype that promotes axonal regeneration. Defining the molecular mechanisms that maintain repair Schwann cells is an essential step toward developing therapeutic strategies that improve nerve regeneration and functional recovery.


Asunto(s)
Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/metabolismo , Fenotipo , Factor de Transcripción STAT3/genética , Células de Schwann/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Ratones , Factor de Transcripción STAT3/metabolismo , Células de Schwann/citología
6.
Blood ; 127(11): 1426-37, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26834239

RESUMEN

The Gata2 transcription factor is a pivotal regulator of hematopoietic cell development and maintenance, highlighted by the fact that Gata2 haploinsufficiency has been identified as the cause of some familial cases of acute myelogenous leukemia/myelodysplastic syndrome and in MonoMac syndrome. Genetic deletion in mice has shown that Gata2 is pivotal to the embryonic generation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). It functions in the embryo during endothelial cell to hematopoietic cell transition to affect hematopoietic cluster, HPC, and HSC formation. Gata2 conditional deletion and overexpression studies show the importance of Gata2 levels in hematopoiesis, during all developmental stages. Although previous studies of cell populations phenotypically enriched in HPCs and HSCs show expression of Gata2, there has been no direct study of Gata2 expressing cells during normal hematopoiesis. In this study, we generate a Gata2Venus reporter mouse model with unperturbed Gata2 expression to examine the hematopoietic function and transcriptome of Gata2 expressing and nonexpressing cells. We show that all the HSCs are Gata2 expressing. However, not all HPCs in the aorta, vitelline and umbilical arteries, and fetal liver require or express Gata2. These Gata2-independent HPCs exhibit a different functional output and genetic program, including Ras and cyclic AMP response element-binding protein pathways and other Gata factors, compared with Gata2-dependent HPCs. Our results, indicating that Gata2 is of major importance in programming toward HSC fate but not in all cells with HPC fate, have implications for current reprogramming strategies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Animales , Aorta/citología , Aorta/embriología , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Linaje de la Célula , Células Cultivadas , Técnicas de Reprogramación Celular , Factor de Transcripción GATA2/deficiencia , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/fisiología , Genes Reporteros , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/clasificación , Células Madre Hematopoyéticas/fisiología , Hígado/citología , Hígado/embriología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transcriptoma , Transgenes , Arterias Umbilicales/citología , Arterias Umbilicales/embriología
7.
Nature ; 491(7425): 554-9, 2012 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-23135404

RESUMEN

Regulatory T (T(reg)) cells, characterized by expression of the transcription factor forkhead box P3 (Foxp3), maintain immune homeostasis by suppressing self-destructive immune responses. Foxp3 operates as a late-acting differentiation factor controlling T(reg) cell homeostasis and function, whereas the early T(reg)-cell-lineage commitment is regulated by the Akt kinase and the forkhead box O (Foxo) family of transcription factors. However, whether Foxo proteins act beyond the T(reg)-cell-commitment stage to control T(reg) cell homeostasis and function remains largely unexplored. Here we show that Foxo1 is a pivotal regulator of T(reg )cell function. T(reg) cells express high amounts of Foxo1 and display reduced T-cell-receptor-induced Akt activation, Foxo1 phosphorylation and Foxo1 nuclear exclusion. Mice with T(reg)-cell-specific deletion of Foxo1 develop a fatal inflammatory disorder similar in severity to that seen in Foxp3-deficient mice, but without the loss of T(reg) cells. Genome-wide analysis of Foxo1 binding sites reveals ~300 Foxo1-bound target genes, including the pro-inflammatory cytokine Ifng, that do not seem to be directly regulated by Foxp3. These findings show that the evolutionarily ancient Akt-Foxo1 signalling module controls a novel genetic program indispensable for T(reg) cell function.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Núcleo Celular/patología , Femenino , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Regulación de la Expresión Génica/genética , Genoma/genética , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Interferón gamma/deficiencia , Interferón gamma/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T Reguladores/patología
8.
Nature ; 485(7399): 517-21, 2012 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-22622581

RESUMEN

Oligodendrocytes, the myelin-forming glial cells of the central nervous system, maintain long-term axonal integrity. However, the underlying support mechanisms are not understood. Here we identify a metabolic component of axon-glia interactions by generating conditional Cox10 (protoheme IX farnesyltransferase) mutant mice, in which oligodendrocytes and Schwann cells fail to assemble stable mitochondrial cytochrome c oxidase (COX, also known as mitochondrial complex IV). In the peripheral nervous system, Cox10 conditional mutants exhibit severe neuropathy with dysmyelination, abnormal Remak bundles, muscle atrophy and paralysis. Notably, perturbing mitochondrial respiration did not cause glial cell death. In the adult central nervous system, we found no signs of demyelination, axonal degeneration or secondary inflammation. Unlike cultured oligodendrocytes, which are sensitive to COX inhibitors, post-myelination oligodendrocytes survive well in the absence of COX activity. More importantly, by in vivo magnetic resonance spectroscopy, brain lactate concentrations in mutants were increased compared with controls, but were detectable only in mice exposed to volatile anaesthetics. This indicates that aerobic glycolysis products derived from oligodendrocytes are rapidly metabolized within white matter tracts. Because myelinated axons can use lactate when energy-deprived, our findings suggest a model in which axon-glia metabolic coupling serves a physiological function.


Asunto(s)
Axones/fisiología , Glucólisis , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Potenciales de Acción , Transferasas Alquil y Aril/deficiencia , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Respiración de la Célula , Supervivencia Celular , Enfermedades Desmielinizantes/enzimología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Ácido Láctico/metabolismo , Espectroscopía de Resonancia Magnética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/enzimología , Protones , Células de Schwann/enzimología , Células de Schwann/metabolismo , Factores de Tiempo
9.
Development ; 141(8): 1749-56, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24715463

RESUMEN

The cellular interactions that drive the formation and maintenance of the insulating myelin sheath around axons are only partially understood. Leucine-rich glioma-inactivated (LGI) proteins play important roles in nervous system development and mutations in their genes have been associated with epilepsy and amyelination. Their function involves interactions with ADAM22 and ADAM23 cell surface receptors, possibly in apposing membranes, thus attenuating cellular interactions. LGI4-ADAM22 interactions are required for axonal sorting and myelination in the developing peripheral nervous system (PNS). Functional analysis revealed that, despite their high homology and affinity for ADAM22, LGI proteins are functionally distinct. To dissect the key residues in LGI proteins required for coordinating axonal sorting and myelination in the developing PNS, we adopted a phylogenetic and computational approach and demonstrate that the mechanism of action of LGI4 depends on a cluster of three amino acids on the outer surface of the LGI4 protein, thus providing a structural basis for the mechanistic differences in LGI protein function in nervous system development and evolution.


Asunto(s)
Glicoproteínas/química , Glicoproteínas/metabolismo , Vaina de Mielina/metabolismo , Filogenia , Proteínas ADAM/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Axones/metabolismo , Secuencia Conservada , Prueba de Complementación Genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos , Sistema Nervioso Periférico/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Relación Estructura-Actividad , Pez Cebra
10.
Development ; 141(7): 1553-61, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24598164

RESUMEN

Myelination allows rapid saltatory propagation of action potentials along the axon and is an essential prerequisite for the normal functioning of the nervous system. During peripheral nervous system (PNS) development, myelin-forming Schwann cells (SCs) generate radial lamellipodia to sort and ensheath axons. This process requires controlled cytoskeletal remodeling, and we show that SC lamellipodia formation depends on the function of profilin 1 (Pfn1), an actin-binding protein involved in microfilament polymerization. Pfn1 is inhibited upon phosphorylation by ROCK, a downstream effector of the integrin linked kinase pathway. Thus, a dramatic reduction of radial lamellipodia formation is observed in SCs lacking integrin-linked kinase or treated with the Rho/ROCK activator lysophosphatidic acid. Knocking down Pfn1 expression by lentiviral-mediated shRNA delivery impairs SC lamellipodia formation in vitro, suggesting a direct role for this protein in PNS myelination. Indeed, SC-specific gene ablation of Pfn1 in mice led to profound radial sorting and myelination defects, confirming a central role for this protein in PNS development. Our data identify Pfn1 as a key effector of the integrin linked kinase/Rho/ROCK pathway. This pathway, acting in parallel with integrin ß1/LCK/Rac1 and their effectors critically regulates SC lamellipodia formation, radial sorting and myelination during peripheral nervous system maturation.


Asunto(s)
Vaina de Mielina/fisiología , Nervios Periféricos/fisiología , Sistema Nervioso Periférico/fisiología , Profilinas/fisiología , Animales , Transporte Axonal/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis/genética , Neuropéptidos/fisiología , Seudópodos/genética , Células de Schwann/fisiología , Proteína de Unión al GTP rac1/fisiología
11.
J Biol Chem ; 290(2): 727-43, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25416780

RESUMEN

The transcription factor Miz1 (Myc-interacting zinc finger 1) is a known regulator of the cell cycle but also has cell cycle-independent functions. Here we analyzed the role of Miz1 in the peripheral nervous system, using an early embryonic conditional knock-out model in which the Miz1 POZ domain is ablated in Schwann cells. Although the development of myelinated nerve fibers was not impaired, Miz1ΔPOZ mice acquired behavioral signs of a peripheral neuropathy at the age of 3 months. At this time, ultrastructural analysis of the sciatic nerve showed de- and dysmyelination of fibers, with massive outfoldings and a focal infiltration of macrophages. Although the expression of genes encoding structural myelin proteins, such as periaxin, myelin basic protein, and myelin protein zero, was decreased, genes associated with a negative regulation of myelination, including c-Jun, Sox2, and Id2, were up-regulated in Miz1ΔPOZ mice compared with controls. In animals older than 4 months, the motor disabilities vanished, and the ultrastructure of the sciatic nerve exhibited numerous tomacula and remyelinated fibers, as indicated by thinner myelin. No second acute attack was observed up to the age of 1 year. Thus, the deletion of the Miz1 POZ domain in Schwann cells induces an acute neuropathy with a subsequent regeneration in which there is ongoing balancing between de- and remyelination. Miz1ΔPOZ mice are impaired in the maintenance of myelinated fibers and are a promising model for studying remyelination in adult peripheral nerves.


Asunto(s)
Regeneración Nerviosa/genética , Proteínas Nucleares/metabolismo , Enfermedades del Sistema Nervioso Periférico/genética , Sistema Nervioso Periférico/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Células de Schwann/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Proteínas Nucleares/genética , Sistema Nervioso Periférico/crecimiento & desarrollo , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/terapia , Proteínas Inhibidoras de STAT Activados/genética , Estructura Terciaria de Proteína/genética , Células de Schwann/patología , Nervio Ciático/metabolismo , Nervio Ciático/patología , Ubiquitina-Proteína Ligasas
12.
Cancer Cell ; 13(2): 105-16, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18242511

RESUMEN

Neurofibromatosis type 1 (Nf1) mutation predisposes to benign peripheral nerve (glial) tumors called neurofibromas. The point(s) in development when Nf1 loss promotes neurofibroma formation are unknown. We show that inactivation of Nf1 in the glial lineage in vitro at embryonic day 12.5 + 1, but not earlier (neural crest) or later (mature Schwann cell), results in colony-forming cells capable of multilineage differentiation. In vivo, inactivation of Nf1 using a DhhCre driver beginning at E12.5 elicits plexiform neurofibromas, dermal neurofibromas, and pigmentation. Tumor Schwann cells uniquely show biallelic Nf1 inactivation. Peripheral nerve and tumors contain transiently proliferating Schwann cells that lose axonal contact, providing insight into early neurofibroma formation. We suggest that timing of Nf1 mutation is critical for neurofibroma formation.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neurofibroma Plexiforme/patología , Neurofibromina 1/metabolismo , Neoplasias del Sistema Nervioso Periférico/patología , Pigmentación , Animales , Axones/metabolismo , Axones/patología , Proliferación Celular , Pérdida del Embrión , Embrión de Mamíferos/citología , Ganglios Espinales/citología , Integrasas/metabolismo , Ratones , Modelos Biológicos , Neurofibroma Plexiforme/ultraestructura , Neuroglía/citología , Neuroglía/metabolismo , Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Recombinación Genética , Células de Schwann/patología , Células de Schwann/ultraestructura , Nervio Ciático/metabolismo , Nervio Ciático/patología , Células Madre/citología , Células Madre/metabolismo
13.
Cell Rep ; 43(1): 113634, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38194969

RESUMEN

Neurodevelopmental disorders, such as intellectual disability (ID), epilepsy, and autism, involve altered synaptic transmission and plasticity. Functional characterization of their associated genes is vital for understanding physio-pathological brain functions. LGI3 is a recently recognized ID-associated gene encoding a secretory protein related to an epilepsy-gene product, LGI1. Here, we find that LGI3 is uniquely secreted from oligodendrocytes in the brain and enriched at juxtaparanodes of myelinated axons, forming nanoscale subclusters. Proteomic analysis using epitope-tagged Lgi3 knockin mice shows that LGI3 uses ADAM23 as a receptor and selectively co-assembles with Kv1 channels. A lack of Lgi3 in mice disrupts juxtaparanodal clustering of ADAM23 and Kv1 channels and suppresses Kv1-channel-mediated short-term synaptic plasticity. Collectively, this study identifies an extracellular organizer of juxtaparanodal Kv1 channel clustering for finely tuned synaptic transmission. Given the defective secretion of the LGI3 missense variant, we propose a molecular pathway, the juxtaparanodal LGI3-ADAM23-Kv1 channel, for understanding neurodevelopmental disorders.


Asunto(s)
Epilepsia , Proteómica , Animales , Ratones , Axones/metabolismo , Epilepsia/metabolismo , Plasticidad Neuronal , Oligodendroglía/metabolismo , Proteínas/metabolismo
14.
J Neurosci ; 32(16): 5472-80, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22514309

RESUMEN

Schwann cells are the main glial cell type in the PNS. They develop along nerves during embryogenesis and rely on the HMG domain containing Sox10 transcription factor for specification, lineage progression, and terminal differentiation. Sox10 deletion in immature Schwann cells caused peripheral nerve defects in mice that were not restricted to this glial cell type, although expression in the nerve and gene loss were. Formation of the perineurium as the protecting sheath was, for instance, heavily compromised. This resembled the defect observed after loss of Desert hedgehog (Dhh) in mice. Here we show that Sox10 activates Dhh expression in Schwann cells via an enhancer that is located in intron 1 of the Dhh gene. Sox10 binds this enhancer in monomeric form via several sites. Mutation of these sites abolishes both Schwann-cell-specific activity and Sox10 responsiveness in vitro and in transgenic mouse embryos. This argues that Sox10 activates Dhh expression by direct binding to the enhancer and by increasing Dhh levels promotes formation of the perineurial sheath. This represents the first mechanism for a non-cell-autonomous function of Sox10 during peripheral nerve development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Hedgehog/metabolismo , Sistema Nervioso Periférico/embriología , Factores de Transcripción SOXE/metabolismo , Animales , Línea Celular Transformada , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Exones/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Hedgehog/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Unión Proteica/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXE/genética , Células de Schwann/metabolismo , Transfección
15.
Brain ; 135(Pt 5): 1395-411, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22451505

RESUMEN

Mutations in dynamin 2 (DNM2) lead to dominant intermediate Charcot-Marie-Tooth neuropathy type B, while a different set of DNM2 mutations cause autosomal dominant centronuclear myopathy. In this study, we aimed to elucidate the disease mechanisms in dominant intermediate Charcot-Marie-Tooth neuropathy type B and to find explanations for the tissue-specific defects that are associated with different DNM2 mutations in dominant intermediate Charcot-Marie-Tooth neuropathy type B versus autosomal dominant centronuclear myopathy. We used tissue derived from Dnm2-deficient mice to establish an appropriate peripheral nerve model and found that dominant intermediate Charcot-Marie-Tooth neuropathy type B-associated dynamin 2 mutants, but not autosomal dominant centronuclear myopathy mutants, impaired myelination. In contrast to autosomal dominant centronuclear myopathy mutants, Schwann cells and neurons from the peripheral nervous system expressing dominant intermediate Charcot-Marie-Tooth neuropathy mutants showed defects in clathrin-mediated endocytosis. We demonstrate that, as a consequence, protein surface levels are altered in Schwann cells. Furthermore, we discovered that myelination is strictly dependent on Dnm2 and clathrin-mediated endocytosis function. Thus, we propose that altered endocytosis is a major contributing factor to the disease mechanisms in dominant intermediate Charcot-Marie-Tooth neuropathy type B.


Asunto(s)
Clatrina/farmacología , Dinamina II/genética , Endocitosis/fisiología , Regulación de la Expresión Génica/genética , Mutación/genética , Neuronas/fisiología , Complejo 2 de Proteína Adaptadora/genética , Complejo 2 de Proteína Adaptadora/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Citometría de Flujo , Ganglios Espinales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Integrina beta1/metabolismo , Ratones , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neuronas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptor ErbB-2/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Factores de Tiempo , Transfección , Transferrina/metabolismo
16.
J Cell Biol ; 222(4)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36828548

RESUMEN

Along myelinated axons, Shaker-type potassium channels (Kv1) accumulate at high density in the juxtaparanodal region, directly adjacent to the paranodal axon-glia junctions that flank the nodes of Ranvier. However, the mechanisms that control the clustering of Kv1 channels, as well as their function at this site, are still poorly understood. Here we demonstrate that axonal ADAM23 is essential for both the accumulation and stability of juxtaparanodal Kv1 complexes. The function of ADAM23 is critically dependent on its interaction with its extracellular ligands LGI2 and LGI3. Furthermore, we demonstrate that juxtaparanodal Kv1 complexes affect the refractory period, thus enabling high-frequency burst firing of action potentials. Our findings not only reveal a previously unknown molecular pathway that regulates Kv1 channel clustering, but they also demonstrate that the juxtaparanodal Kv1 channels that are concealed below the myelin sheath, play a significant role in modifying axonal physiology.


Asunto(s)
Proteínas ADAM , Axones , Vaina de Mielina , Proteínas del Tejido Nervioso , Canales de Potasio con Entrada de Voltaje , Potenciales de Acción , Axones/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Vaina de Mielina/metabolismo , Neuroglía/metabolismo , Nódulos de Ranvier/metabolismo , Proteínas ADAM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo
17.
J Neurosci ; 31(8): 3032-43, 2011 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-21414924

RESUMEN

Myelination is dependent on complex reciprocal interactions between the Schwann cell (SC) and axon. Recent evidence suggests that the SC-axon interface represents a membrane specialization essential for myelination; however, the manner in which this polarized-apical domain is generated remains a mystery. The cell adhesion molecule N-cadherin is enriched at the SC-axon interface and colocalizes with the polarity protein Par-3. The asymmetric localization is induced on SC-SC and SC-axon contact. Knockdown of N-cadherin in SCs cocultured with DRG neurons disrupts Par-3 localization and delays the initiation of myelination. However, knockdown or overexpression of neuronal N-cadherin does not influence the distribution of Par-3 or myelination, suggesting that homotypic interactions between SC and axonal N-cadherin are not essential for the events surrounding myelination. To further investigate the role of N-cadherin, mice displaying SC-specific gene ablation of N-cadherin were generated and characterized. Surprisingly, myelination is only slightly delayed, and mice are viable without any detectable myelination defects. ß-Catenin, a downstream effector of N-cadherin, colocalizes and coimmunoprecipitates with N-cadherin on the initiation of myelination. To determine whether ß-catenin mediates compensation on N-cadherin deletion, SC-specific gene ablation of ß-catenin was generated and characterized. Consistent with our hypothesis, myelination is more severely delayed than when manipulating N-cadherin alone, but without any defect to the myelin sheath. Together, our results suggest that N-cadherin interacts with ß-catenin in establishing SC polarity and the timely initiation of myelination, but they are nonessential components for the formation and maturation of the myelin sheath.


Asunto(s)
Axones/fisiología , Cadherinas/fisiología , Ganglios Espinales/embriología , Vaina de Mielina/fisiología , Células de Schwann/metabolismo , beta Catenina/fisiología , Animales , Animales Recién Nacidos , Cadherinas/genética , Polaridad Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Adhesiones Focales/fisiología , Ganglios Espinales/citología , Ganglios Espinales/crecimiento & desarrollo , Ratones , Ratones Noqueados , Ratas , Células de Schwann/citología , Células de Schwann/fisiología , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , beta Catenina/genética
18.
J Neurosci ; 31(23): 8585-94, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21653862

RESUMEN

The POU domain transcription factor Pou3f1 (Oct6/Scip/Tst1) initiates the transition from ensheathing, promyelinating Schwann cells to myelinating cells. Axonal and other extracellular signals regulate Oct6 expression through the Oct6 Schwann cell enhancer (SCE), which is both required and sufficient to drive all aspects of Oct6 expression in Schwann cells. Thus, the Oct6 SCE is pivotal in the gene regulatory network that governs the onset of myelin formation in Schwann cells and provides a link between myelin promoting signaling and activation of a myelin-related transcriptional network. In this study, we define the relevant cis-acting elements within the SCE and identify the transcription factors that mediate Oct6 regulation. On the basis of phylogenetic comparisons and functional in vivo assays, we identify a number of highly conserved core elements within the mouse SCE. We show that core element 1 is absolutely required for full enhancer function and that it contains closely spaced inverted binding sites for Sox proteins. For the first time in vivo, the dimeric Sox10 binding to this element is shown to be essential for enhancer activity, whereas monomeric Sox10 binding is nonfunctional. As Oct6 and Sox10 synergize to activate the expression of the major myelin-related transcription factor Krox20, we propose that Sox10-dependent activation of Oct6 defines a feedforward regulatory module that serves to time and amplify the onset of myelination in the peripheral nervous system.


Asunto(s)
Vaina de Mielina/metabolismo , Factor 6 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXE/metabolismo , Células de Schwann/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Unión Proteica , Ratas , Células de Schwann/citología
19.
Am J Respir Cell Mol Biol ; 46(2): 224-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22298531

RESUMEN

Alveolarization of the developing lung is an important step toward the switch from intrauterine life to breathing oxygen-rich air after birth. The distal airways structurally change to minimize the gas exchange path, and Type II pneumocytes increase the production of surfactants, which are required to reduce surface tension at the air-liquid interface in the alveolus. Hypoxia-inducible factor 2α (Hif2α) is an oxygen-regulated transcription factor expressed in endothelial and Type II cells, and its expression increases toward the end of gestation. We investigated the role of Hif2α in Type II cells by conditionally expressing an oxygen-insensitive mutant of Hif2α in airway epithelial cells during development. Newborn mice expressing the mutant Hif2α were born alive but quickly succumbed to respiratory distress. Subsequent analysis of the lungs revealed dilated alveoli covered with enlarged, aberrant Type II cells and a diminished number of Type I cells. The Type II cells accumulated glycogen in part by increased glucose uptake via the up-regulation of the glucose transporter 1. Furthermore, the cells lacked two crucial enzymes involved in the metabolism of glycogen into surfactant lipids, lysophosphatidylcholine acyltransferase and ATP-binding cassette sub-family A member 3. We conclude that Hif2α is a key regulator in alveolar maturation and the production of phospholipids by Type II cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Alveolos Pulmonares/fisiopatología , Surfactantes Pulmonares , Animales , Humanos , Ratones
20.
J Biol Chem ; 286(30): 26781-93, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21715287

RESUMEN

The Lpin1 gene encodes the phosphatidate phosphatase (PAP1) enzyme Lipin 1, which plays a critical role in lipid metabolism. In this study we describe the identification and characterization of a rat model with a mutated Lpin1 gene (Lpin1(1Hubr)), generated by N-ethyl-N-nitrosourea mutagenesis. Lpin1(1Hubr) rats are characterized by hindlimb paralysis and mild lipodystrophy that are detectable from the second postnatal week. Sequencing of Lpin1 identified a point mutation in the 5'-end splice site of intron 18 resulting in mis-splicing, a reading frameshift, and a premature stop codon. As this mutation does not induce nonsense-mediated decay, it allows the production of a truncated Lipin 1 protein lacking PAP1 activity. Lpin1(1Hubr) rats developed hypomyelination and mild lipodystrophy rather than the pronounced demyelination and adipocyte defects characteristic of Lpin1(fld/fld) mice, which carry a null allele for Lpin1. Furthermore, biochemical, histological, and molecular analyses revealed that these lesions improve in older Lpin1(1Hubr) rats as compared with young Lpin1(1Hubr) rats and Lpin1(fld/fld) mice. We observed activation of compensatory biochemical pathways substituting for missing PAP1 activity that, in combination with a possible non-enzymatic Lipin 1 function residing outside of its PAP1 domain, may contribute to the less severe phenotypes observed in Lpin1(1Hubr) rats as compared with Lpin1(fld/fld) mice. Although we are cautious in making a direct parallel between the presented rodent model and human disease, our data may provide new insight into the pathogenicity of recently identified human LPIN1 mutations.


Asunto(s)
Enfermedades Desmielinizantes/enzimología , Intrones , Lipodistrofia/enzimología , Mutación , Fosfatidato Fosfatasa/metabolismo , Alquilantes/farmacología , Animales , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Etilnitrosourea/farmacología , Células HEK293 , Humanos , Lipodistrofia/genética , Lipodistrofia/patología , Ratones , Mutagénesis , Proteínas Asociadas a Pancreatitis , Fosfatidato Fosfatasa/genética , Estructura Terciaria de Proteína , Sitios de Empalme de ARN , Ratas , Ratas Mutantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA