Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39131270

RESUMEN

Docosahexaenoic acid (DHA), a dietary omega-3 fatty acid, is a major building block of brain cell membranes. Offspring rely on maternal DHA transfer to meet their neurodevelopmental needs, but DHA sources are lacking in the American diet. Low DHA status is linked to altered immune responses, white matter defects, impaired vision, and an increased risk of psychiatric disorders during development. However, the underlying cellular mechanisms involved are largely unknown, and advancements in the field have been limited by the existing tools and animal models. Zebrafish are an excellent model for studying neurodevelopmental mechanisms. Embryos undergo rapid external development and are optically transparent, enabling direct observation of individual cells and dynamic cell-cell interactions in a way that is not possible in rodents. Here, we create a novel DHA-deficient zebrafish model by 1) disrupting elovl2, a key gene in the DHA biosynthesis pathway, via CRISPR-Cas9 genome editing, and 2) feeding mothers a DHA-deficient diet. We show that low DHA status during development is associated with a small eye morphological phenotype and demonstrate that even the morphologically normal siblings exhibit dysregulated gene pathways related to vision and stress response. Future work using our zebrafish model could reveal the cellular and molecular mechanisms by which low DHA status leads to neurodevelopmental abnormalities and provide insight into maternal nutritional strategies that optimize infant brain health.

2.
bioRxiv ; 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39149260

RESUMEN

The extracellular matrix (ECM) provides critical biochemical and structural cues that regulate neural development. Chondroitin sulfate proteoglycans (CSPGs), a major ECM component, have been implicated in modulating oligodendrocyte precursor cell (OPC) proliferation, migration, and maturation, but their specific roles in oligodendrocyte lineage cell (OLC) development and myelination in vivo remain poorly understood. Here, we use zebrafish as a model system to investigate the spatiotemporal dynamics of ECM deposition and CSPG localization during central nervous system (CNS) development, with a focus on their relationship to OLCs. We demonstrate that ECM components, including CSPGs, are dynamically expressed in distinct spatiotemporal patterns coinciding with OLC development and myelination. We found that zebrafish lacking cspg4 function produced normal numbers of OLCs, which appeared to undergo proper differentiation. However, OPC morphology in mutant larvae was aberrant. Nevertheless, the number and length of myelin sheaths produced by mature oligodendrocytes were unaffected. These data indicate that Cspg4 regulates OPC morphogenesis in vivo, supporting the role of the ECM in neural development.

3.
J Neurosci ; 44(31)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-38969506

RESUMEN

Although hyperactivity is associated with a wide variety of neurodevelopmental disorders, the early embryonic origins of locomotion have hindered investigation of pathogenesis of these debilitating behaviors. The earliest motor output in vertebrate animals is generated by clusters of early-born motor neurons (MNs) that occupy distinct regions of the spinal cord, innervating stereotyped muscle groups. Gap junction electrical synapses drive early spontaneous behavior in zebrafish, prior to the emergence of chemical neurotransmitter networks. We use a genetic model of hyperactivity to gain critical insight into the consequences of errors in motor circuit formation and function, finding that Fragile X syndrome model mutant zebrafish are hyperexcitable from the earliest phases of spontaneous behavior, show altered sensitivity to blockade of electrical gap junctions, and have increased expression of the gap junction protein Connexin 34/35. We further show that this hyperexcitable behavior can be rescued by pharmacological inhibition of electrical synapses. We also use functional imaging to examine MN and interneuron (IN) activity in early embryogenesis, finding genetic disruption of electrical gap junctions uncouples activity between mnx1 + MNs and INs. Taken together, our work highlights the importance of electrical synapses in motor development and suggests that the origins of hyperactivity in neurodevelopmental disorders may be established during the initial formation of locomotive circuits.


Asunto(s)
Sinapsis Eléctricas , Síndrome del Cromosoma X Frágil , Neuronas Motoras , Proteínas de Pez Cebra , Pez Cebra , Animales , Síndrome del Cromosoma X Frágil/fisiopatología , Síndrome del Cromosoma X Frágil/genética , Sinapsis Eléctricas/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Neuronas Motoras/fisiología , Modelos Animales de Enfermedad , Conexinas/genética , Conexinas/metabolismo , Animales Modificados Genéticamente , Hipercinesia/fisiopatología , Interneuronas/fisiología , Interneuronas/metabolismo , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo
4.
Front Endocrinol (Lausanne) ; 13: 1033843, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36578958

RESUMEN

The Notch pathway is a cell-cell communication system which is critical for many developmental processes, including craniofacial development. Notch receptor activation induces expression of several well-known canonical targets including those encoded by the hes and her genes in mammals and zebrafish, respectively. The function of these genes, individually and in combination, during craniofacial development is not well understood. Here, we used zebrafish genetics to investigate her9 and her6 gene function during craniofacial development. We found that her9 is required for osteoblasts to efficiently mineralize bone, while cartilage is largely unaffected. Strikingly, gene expression studies in her9 mutants indicate that although progenitor cells differentiate into osteoblasts at the appropriate time and place, they fail to efficiently lay down mineralized matrix. This mineralization role of her9 is likely independent of Notch activation. In contrast, her9 also functions redundantly with her6 downstream of Jagged1b-induced Notch activation during dorsoventral craniofacial patterning. These studies disentangle distinct and redundant her gene functions during craniofacial development, including an unexpected, Notch independent, requirement during bone mineralization.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Receptores Notch/genética , Huesos/metabolismo , Mamíferos/metabolismo
5.
J Neurosci ; 41(41): 8532-8544, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34475201

RESUMEN

In the vertebrate CNS, oligodendrocytes produce myelin, a specialized membrane, to insulate and support axons. Individual oligodendrocytes wrap multiple axons with myelin sheaths of variable lengths and thicknesses. Myelin grows at the distal ends of oligodendrocyte processes, and multiple lines of work have provided evidence that mRNAs and RNA binding proteins localize to myelin, together supporting a model where local translation controls myelin sheath growth. What signal transduction mechanisms could control this? One strong candidate is the Akt-mTOR pathway, a major cellular signaling hub that coordinates transcription, translation, metabolism, and cytoskeletal organization. Here, using zebrafish as a model system, we found that Akt-mTOR signaling promotes myelin sheath growth and stability during development. Through cell-specific manipulations to oligodendrocytes, we show that the Akt-mTOR pathway drives cap-dependent translation to promote myelination and that restoration of cap-dependent translation is sufficient to rescue myelin deficits in mTOR loss-of-function animals. Moreover, an mTOR-dependent translational regulator was phosphorylated and colocalized with mRNA encoding a canonically myelin-translated protein in vivo, and bioinformatic investigation revealed numerous putative translational targets in the myelin transcriptome. Together, these data raise the possibility that Akt-mTOR signaling in nascent myelin sheaths promotes sheath growth via translation of myelin-resident mRNAs during development.SIGNIFICANCE STATEMENT In the brain and spinal cord, oligodendrocytes extend processes that tightly wrap axons with myelin, a protein- and lipid-rich membrane that increases electrical impulses and provides trophic support. Myelin membrane grows dramatically following initial axon wrapping in a process that demands protein and lipid synthesis. How protein and lipid synthesis is coordinated with the need for myelin to be generated in certain locations remains unknown. Our study reveals that the Akt-mTOR signaling pathway promotes myelin sheath growth by regulating protein translation. Because we found translational regulators of the Akt-mTOR pathway in myelin, our data raise the possibility that Akt-mTOR activity regulates translation in myelin sheaths to deliver myelin on demand to the places it is needed.


Asunto(s)
Vaina de Mielina/metabolismo , Biosíntesis de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Caperuzas de ARN/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Femenino , Humanos , Masculino , Vaina de Mielina/genética , Proteínas Proto-Oncogénicas c-akt/genética , Caperuzas de ARN/genética , Serina-Treonina Quinasas TOR/genética , Pez Cebra , Proteínas de Pez Cebra/genética
6.
Dev Biol ; 479: 37-50, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34303700

RESUMEN

Ventral spinal cord progenitor cells, which express the basic helix loop helix transcription factor Olig2, sequentially produce motor neurons and oligodendrocyte precursor cells (OPCs). Following specification some OPCs differentiate as myelinating oligodendrocytes while others persist as OPCs. Though a considerable amount of work has described the molecular profiles that define motor neurons, OPCs, and oligodendrocytes, less is known about the progenitors that produce them. To identify the developmental origins and transcriptional profiles of motor neurons and OPCs, we performed single-cell RNA sequencing on isolated pMN cells from embryonic zebrafish trunk tissue at stages that encompassed motor neurogenesis, OPC specification, and initiation of oligodendrocyte differentiation. Downstream analyses revealed two distinct pMN progenitor populations: one that appears to produce neurons and one that appears to produce OPCs. This latter population, called Pre-OPCs, is marked by expression of GS Homeobox 2 (gsx2), a gene that encodes a homeobox transcription factor. Using fluorescent in situ hybridizations, we identified gsx2-expressing Pre-OPCs in the spinal cord prior to expression of canonical OPC marker genes. Our data therefore reveal heterogeneous gene expression profiles among pMN progenitors, supporting prior fate mapping evidence.


Asunto(s)
Diferenciación Celular/fisiología , Células-Madre Neurales/citología , Médula Espinal/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linaje de la Célula , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Hedgehog/metabolismo , Neuronas Motoras/citología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Oligodendroglía/citología , Análisis de la Célula Individual/métodos , Análisis Espacio-Temporal , Factores de Transcripción/metabolismo , Transcriptoma/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
7.
Glia ; 69(10): 2349-2361, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34110049

RESUMEN

Neurodevelopment requires the precise integration of a wide variety of neuronal and glial cell types. During early embryonic development, motor neurons and then oligodendrocyte precursor cells (OPCs) are specified from neural progenitors residing in the periventricular pMN progenitor domain of the spinal cord. Following gliogenesis, OPCs can differentiate as oligodendrocytes (OLs)-the myelinating glial cells of the central nervous system-or remain as OPCs. To generate unique cell types capable of highly divergent functions, these specification and differentiation events require specialized gene expression programs. RNA binding proteins (RBPs) regulate mRNA localization and translation in the developing nervous system and are linked to many neurodevelopmental disorders. One example is Fragile X syndrome (FXS), caused by the loss of the RBP fragile X mental retardation protein (FMRP). Importantly, infants with FXS have reduced white matter and we previously showed that zebrafish Fmrp is autonomously required in OLs to promote myelin sheath growth. We now find that Fmrp regulates cell specification in pMN progenitor cells such that fmr1 mutant zebrafish generate fewer motor neurons and excess OPCs. Fmrp subsequently promotes differentiation of OPCs, leading to fewer differentiating OLs in the developing spinal cord of fmr1 larvae. Although the early patterning of spinal progenitor domains appears largely normal in fmr1 mutants during early embryogenesis, Shh signaling is greatly diminished. Taken together, these results suggest cell stage-specific requirements for Fmrp in the specification and differentiation of oligodendrocyte lineage cells.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Proteínas de Unión al ARN , Proteínas de Pez Cebra , Pez Cebra , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Neuronas Motoras/metabolismo , Oligodendroglía/metabolismo , Embarazo , Pez Cebra/metabolismo
8.
Genetics ; 218(4)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34057474

RESUMEN

The axis of the vertebrate neural tube is patterned, in part, by a ventral to dorsal gradient of Shh signaling. In the ventral spinal cord, Shh induces concentration-dependent expression of transcription factors, subdividing neural progenitors into distinct domains that subsequently produce distinct neuronal and glial subtypes. In particular, progenitors of the pMN domain express the bHLH transcription factor Olig2 and produce motor neurons followed by oligodendrocytes, the myelinating glial cell type of the central nervous system. In addition to its role in patterning ventral progenitors, Shh signaling must be maintained through development to specify pMN progenitors for oligodendrocyte fate. Using a forward genetic screen in zebrafish for mutations that disrupt the development of oligodendrocytes, we identified a new mutant allele of boc, which encodes a type I transmembrane protein that functions as a coreceptor for Shh. Embryos homozygous for the bocco25 allele, which creates a missense mutation in a Fibronectin type III domain that binds Shh, have normally patterned spinal cords but fail to maintain pMN progenitors, resulting in a deficit of oligodendrocytes. Using a sensitive fluorescent detection method for in situ RNA hybridization, we found that spinal cord cells express boc in a graded fashion that is inverse to the gradient of Shh signaling activity and that boc function is necessary to maintain pMN progenitors by shaping the Shh signaling gradient.


Asunto(s)
Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neurogénesis , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Moléculas de Adhesión de Célula Nerviosa/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Oligodendroglía/citología , Médula Espinal/citología , Médula Espinal/embriología , Pez Cebra , Proteínas de Pez Cebra/genética
9.
PLoS Biol ; 19(1): e3001053, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33439856

RESUMEN

Myelin is a specialized membrane produced by oligodendrocytes that insulates and supports axons. Oligodendrocytes extend numerous cellular processes, as projections of the plasma membrane, and simultaneously wrap multiple layers of myelin membrane around target axons. Notably, myelin sheaths originating from the same oligodendrocyte are variable in size, suggesting local mechanisms regulate myelin sheath growth. Purified myelin contains ribosomes and hundreds of mRNAs, supporting a model that mRNA localization and local protein synthesis regulate sheath growth and maturation. However, the mechanisms by which mRNAs are selectively enriched in myelin sheaths are unclear. To investigate how mRNAs are targeted to myelin sheaths, we tested the hypothesis that transcripts are selected for myelin enrichment through consensus sequences in the 3' untranslated region (3' UTR). Using methods to visualize mRNA in living zebrafish larvae, we identified candidate 3' UTRs that were sufficient to localize mRNA to sheaths and enriched near growth zones of nascent membrane. We bioinformatically identified motifs common in 3' UTRs from 3 myelin-enriched transcripts and determined that these motifs are required and sufficient in a context-dependent manner for mRNA transport to myelin sheaths. Finally, we show that 1 motif is highly enriched in the myelin transcriptome, suggesting that this sequence is a global regulator of mRNA localization during developmental myelination.


Asunto(s)
Regiones no Traducidas 3'/fisiología , Vaina de Mielina/metabolismo , Transporte de ARN/genética , ARN Mensajero/metabolismo , Animales , Animales Modificados Genéticamente , Embrión no Mamífero , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Imagen Molecular/métodos , Vaina de Mielina/genética , ARN Mensajero/química , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico/fisiología , Análisis de Secuencia de ARN , Distribución Tisular , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Hum Mutat ; 42(4): 392-407, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33382518

RESUMEN

Idiopathic scoliosis (IS) is a spinal disorder affecting up to 3% of otherwise healthy children. IS has a strong familial genetic component and is believed to be genetically complex due to significant variability in phenotype and heritability. Previous studies identified putative loci and variants possibly contributing to IS susceptibility, including within extracellular matrix, cilia, and actin networks, but the genetic architecture and underlying mechanisms remain unresolved. Here, we used whole-exome sequencing from three affected individuals in a multigenerational family with IS and identified 19 uncommon variants (minor allele frequency < 0.05). Genotyping of additional family members identified a candidate heterozygous variant (H1115Q, G>C, rs142032413) within the ciliary gene KIF7, a regulator within the hedgehog (Hh) signaling pathway. Resequencing of the second cohort of unrelated IS individuals and controls identified several severe mutations in KIF7 in affected individuals only. Subsequently, we generated a mutant zebrafish model of kif7 using CRISPR-Cas9. kif7co63/co63 zebrafish displayed severe scoliosis, presenting in juveniles and progressing through adulthood. We observed no deformities in the brain, Reissner fiber, or central canal cilia in kif7co63/co63 embryos, although alterations were seen in Hh pathway gene expression. This study suggests defects in KIF7-dependent Hh signaling, which may drive pathogenesis in a subset of individuals with IS.


Asunto(s)
Cinesinas , Escoliosis , Pez Cebra , Animales , Cilios/metabolismo , Humanos , Cinesinas/genética , Mutación , Escoliosis/genética , Pez Cebra/genética , Proteínas de Pez Cebra
11.
Biol Open ; 9(10)2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32973079

RESUMEN

The Rab11 apical recycling endosome pathway is a well-established regulator of polarity and lumen formation; however, Rab11-vesicular trafficking also directs a diverse array of other cellular processes, raising the question of how Rab11 vesicles achieve specificity in space, time and content of cargo delivery. In part, this specificity is achieved through effector proteins, yet the role of Rab11 effector proteins in vivo remains vague. Here, we use CRISPR/Cas9 gene editing to study the role of the Rab11 effector Fip5 during zebrafish intestinal development. Zebrafish contain two paralogous genes, fip5a and fip5b, that are orthologs of human FIP5 We find that fip5a- and fip5b-mutant fish show phenotypes characteristic of microvillus inclusion disease, including microvilli defects and lysosomal accumulation. Single and double mutant analyses suggest that fip5a and fip5b function in parallel and regulate trafficking pathways required for assembly of keratin at the terminal web. Remarkably, in some genetic backgrounds, the absence of Fip5 triggers protein upregulation of a closely related family member, Fip1. This compensation mechanism occurs both during zebrafish intestinal development and in tissue culture models of lumenogenesis. In conclusion, our data implicate the Rab11 effectors Fip5 and Fip1 in a trafficking pathway required for apical microvilli formation.


Asunto(s)
Proteínas Portadoras/metabolismo , Intestinos/embriología , Organogénesis/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Unión al GTP rab/metabolismo , Animales , Proteínas Portadoras/genética , Endosomas , Microvellosidades/genética , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Mutación , Fenotipo , Unión Proteica
12.
Development ; 147(16)2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32680935

RESUMEN

Spinal cord pMN progenitors sequentially produce motor neurons and oligodendrocyte precursor cells (OPCs). Some OPCs differentiate rapidly as myelinating oligodendrocytes, whereas others remain into adulthood. How pMN progenitors switch from producing motor neurons to OPCs with distinct fates is poorly understood. pMN progenitors express prdm8, which encodes a transcriptional repressor, during motor neuron and OPC formation. To determine whether prdm8 controls pMN cell fate specification, we used zebrafish as a model system to investigate prdm8 function. Our analysis revealed that prdm8 mutant embryos have fewer motor neurons resulting from a premature switch from motor neuron to OPC production. Additionally, prdm8 mutant larvae have excess oligodendrocytes and a concomitant deficit of OPCs. Notably, pMN cells of mutant embryos have elevated Shh signaling, coincident with the motor neuron to OPC switch. Inhibition of Shh signaling restored the number of motor neurons to normal but did not rescue the proportion of oligodendrocytes. These data suggest that Prdm8 regulates the motor neuron-OPC switch by controlling the level of Shh activity in pMN progenitors, and also regulates the allocation of oligodendrocyte lineage cell fates.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas Hedgehog/metabolismo , Histona Metiltransferasas/metabolismo , Neuronas Motoras/metabolismo , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal , Animales , Proteínas de Unión al ADN/genética , Proteínas Hedgehog/genética , Histona Metiltransferasas/genética , Ratones , Ratones Transgénicos , Neuronas Motoras/citología , Células-Madre Neurales/citología , Oligodendroglía/citología
13.
Nat Neurosci ; 23(9): 1055-1066, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32632287

RESUMEN

During development, oligodendrocytes contact and wrap neuronal axons with myelin. Similarly to neurons and synapses, excess myelin sheaths are produced and selectively eliminated, but how elimination occurs is unknown. Microglia, the resident immune cells of the central nervous system, engulf surplus neurons and synapses. To determine whether microglia also prune myelin sheaths, we used zebrafish to visualize and manipulate interactions between microglia, oligodendrocytes, and neurons during development. We found that microglia closely associate with oligodendrocytes and specifically phagocytose myelin sheaths. By using a combination of optical, genetic, chemogenetic, and behavioral approaches, we reveal that neuronal activity bidirectionally balances microglial association with neuronal cell bodies and myelin phagocytosis in the optic tectum. Furthermore, multiple strategies to deplete microglia resulted in oligodendrocytes maintaining excessive and ectopic myelin. Our work reveals a neuronal activity-regulated role for microglia in modifying developmental myelin targeting by oligodendrocytes.


Asunto(s)
Vaina de Mielina/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Oligodendroglía/fisiología , Fagocitosis/fisiología , Animales , Animales Modificados Genéticamente , Microglía , Médula Espinal/embriología , Colículos Superiores/embriología , Pez Cebra
14.
Glia ; 68(3): 495-508, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31626382

RESUMEN

During development, oligodendrocytes in the central nervous system extend a multitude of processes that wrap axons with myelin. The highly polarized oligodendrocytes generate myelin sheaths on many different axons, which are far removed from the cell body. Neurons use RNA binding proteins to transport, stabilize, and locally translate mRNA in distal domains of neurons. Local synthesis of synaptic proteins during neurodevelopment facilitates the rapid structural and functional changes underlying neural plasticity and avoids extensive protein transport. We hypothesize that RNA binding proteins also regulate local mRNA regulation in oligodendrocytes to promote myelin sheath growth. Fragile X mental retardation protein (FMRP), an RNA binding protein that plays essential roles in the growth and maturation of neurons, is also expressed in oligodendrocytes. To determine whether oligodendrocytes require FMRP for myelin sheath development, we examined fmr1-/- mutant zebrafish and drove FMR1 expression specifically in oligodendrocytes. We found oligodendrocytes in fmr1-/- mutants developed myelin sheaths of diminished length, a phenotype that can be autonomously rescued in oligodendrocytes with FMR1 expression. Myelin basic protein (Mbp), an essential myelin protein, was reduced in myelin tracts of fmr1-/- mutants, but loss of FMRP function did not impact the localization of mbpa transcript in myelin. Finally, expression of FMR1-I304N, a missense allele that abrogates FMRP association with ribosomes, failed to rescue fmr1-/- mutant sheath growth and induced short myelin sheaths in oligodendrocytes of wild-type larvae. Taken together, these data suggest that FMRP promotes sheath growth through local regulation of translation.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Vaina de Mielina/genética , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , ARN Mensajero/metabolismo , Pez Cebra
15.
Nat Commun ; 10(1): 4125, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511515

RESUMEN

Vesicular release from neurons promotes myelin sheath growth on axons. Oligodendrocytes express proteins that allow dendrites to respond to vesicular release at synapses, suggesting that axon-myelin contacts use similar communication mechanisms as synapses to form myelin sheaths. To test this, we used fusion proteins to track synaptic vesicle localization and membrane fusion in zebrafish during developmental myelination and investigated expression and localization of PSD95, a dendritic post-synaptic protein, within oligodendrocytes. Synaptic vesicles accumulate and exocytose at ensheathment sites with variable patterning and most sheaths localize PSD95 with patterning similar to exocytosis site location. Disruption of candidate PDZ-binding transsynaptic adhesion proteins in oligodendrocytes cause variable effects on sheath length and number. One candidate, Cadm1b, localizes to myelin sheaths where both PDZ binding and extracellular adhesion to axons mediate sheath growth. Our work raises the possibility that axon-glial communication contributes to myelin plasticity, providing new targets for mechanistic unraveling of developmental myelination.


Asunto(s)
Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Sinapsis/metabolismo , Animales , Axones/metabolismo , Moléculas de Adhesión Celular/metabolismo , Exocitosis , Modelos Biológicos , Proteínas del Tejido Nervioso , Oligodendroglía/citología , Vesículas Sinápticas/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra
16.
Dev Biol ; 444(2): 93-106, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30347186

RESUMEN

During development of the central nervous system oligodendrocyte precursor cells (OPCs) give rise to both myelinating oligodendrocytes and NG2 glia, which are the most proliferative cells in the adult mammalian brain. NG2 glia retain characteristics of OPCs, and some NG2 glia produce oligodendrocytes, but many others persist throughout adulthood. Why some OPCs differentiate as oligodendrocytes during development whereas others persist as OPCs and acquire characteristics of NG2 glia is not known. Using zebrafish spinal cord as a model, we found that OPCs that differentiate rapidly as oligodendrocytes and others that remain as OPCs arise in sequential waves from distinct neural progenitors. Additionally, oligodendrocyte and persistent OPC fates are specified during a defined critical period by small differences in Shh signaling and Notch activity, which modulates Shh signaling response. Thus, our data indicate that OPCs fated to produce oligodendrocytes or remain as OPCs during development are specified as distinct cell types, raising the possibility that the myelinating potential of OPCs is set by graded Shh signaling activity.


Asunto(s)
Proteínas Hedgehog/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Receptores Notch/metabolismo , Animales , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula , Proliferación Celular , Sistema Nervioso Central/metabolismo , Neuroglía/metabolismo , Células Precursoras de Oligodendrocitos/fisiología , Oligodendroglía/fisiología , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Células Madre/metabolismo , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
Hum Mol Genet ; 26(15): 2838-2849, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28449119

RESUMEN

CblX (MIM309541) is an X-linked recessive disorder characterized by defects in cobalamin (vitamin B12) metabolism and other developmental defects. Mutations in HCFC1, a transcriptional co-regulator which interacts with multiple transcription factors, have been associated with cblX. HCFC1 regulates cobalamin metabolism via the regulation of MMACHC expression through its interaction with THAP11, a THAP domain-containing transcription factor. The HCFC1/THAP11 complex potentially regulates genes involved in diverse cellular functions including cell cycle, proliferation, and transcription. Thus, it is likely that mutation of THAP11 also results in biochemical and other phenotypes similar to those observed in patients with cblX. We report a patient who presented with clinical and biochemical phenotypic features that overlap cblX, but who does not have any mutations in either MMACHC or HCFC1. We sequenced THAP11 by Sanger sequencing and discovered a potentially pathogenic, homozygous variant, c.240C > G (p.Phe80Leu). Functional analysis in the developing zebrafish embryo demonstrated that both THAP11 and HCFC1 regulate the proliferation and differentiation of neural precursors, suggesting important roles in normal brain development. The loss of THAP11 in zebrafish embryos results in craniofacial abnormalities including the complete loss of Meckel's cartilage, the ceratohyal, and all of the ceratobranchial cartilages. These data are consistent with our previous work that demonstrated a role for HCFC1 in vertebrate craniofacial development. High throughput RNA-sequencing analysis reveals several overlapping gene targets of HCFC1 and THAP11. Thus, both HCFC1 and THAP11 play important roles in the regulation of cobalamin metabolism as well as other pathways involved in early vertebrate development.


Asunto(s)
Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Vitamina B 12/metabolismo , Animales , Secuencia de Bases , Región Branquial/metabolismo , Diferenciación Celular , Niño , Anomalías Craneofaciales/genética , Fibroblastos , Regulación de la Expresión Génica/genética , Factor C1 de la Célula Huésped/química , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Mutación , Cultivo Primario de Células , Transcripción Genética , Vitamina B 12/genética , Pez Cebra/genética
18.
J Neurosci ; 36(29): 7628-39, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27445141

RESUMEN

UNLABELLED: Myelin, which ensheaths and insulates axons, is a specialized membrane highly enriched with cholesterol. During myelin formation, cholesterol influences membrane fluidity, associates with myelin proteins such as myelin proteolipid protein, and assembles lipid-rich microdomains within membranes. Surprisingly, cholesterol also is required by oligodendrocytes, glial cells that make myelin, to express myelin genes and wrap axons. How cholesterol mediates these distinct features of oligodendrocyte development is not known. One possibility is that cholesterol promotes myelination by facilitating signal transduction within the cell, because lipid-rich microdomains function as assembly points for signaling molecules. Signaling cascades that localize to cholesterol-rich regions of the plasma membrane include the PI3K/Akt pathway, which acts upstream of mechanistic target of rapamycin (mTOR), a major driver of myelination. Through manipulation of cholesterol levels and PI3K/Akt/mTOR signaling in zebrafish, we discovered that mTOR kinase activity in oligodendrocytes requires cholesterol. Drawing on a combination of pharmacological and rescue experiments, we provide evidence that mTOR kinase activity is required for cholesterol-mediated myelin gene expression. On the other hand, cholesterol-dependent axon ensheathment is mediated by Akt signaling, independent of mTOR kinase activity. Our data reveal that cholesterol-dependent myelin gene expression and axon ensheathment are facilitated by distinct signaling cascades downstream of Akt. Because mTOR promotes cholesterol synthesis, our data raise the possibility that cholesterol synthesis and mTOR signaling engage in positive feedback to promote the formation of myelin membrane. SIGNIFICANCE STATEMENT: The speed of electrical impulse movement through axons is increased by myelin, a specialized, cholesterol-rich glial cell membrane that tightly wraps axons. During development, myelin membrane grows dramatically, suggesting a significant demand on mechanisms that produce and assemble myelin components, while it spirally wraps axons. Our studies indicate that cholesterol is necessary for both myelin growth and axon wrapping. Specifically, we found that cholesterol facilitates signaling mediated by the PI3K/Akt/mTOR pathway, a powerful driver of myelination. Because mTOR promotes the expression of genes necessary for cholesterol synthesis, cholesterol formation and PI3K/Akt/mTOR signaling might function as a feedforward mechanism to produce the large amounts of myelin membrane necessary for axon ensheathment.


Asunto(s)
Axones/fisiología , Colesterol/biosíntesis , Regulación de la Expresión Génica/fisiología , Proteínas de la Mielina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Animales Modificados Genéticamente , Colesterol/farmacología , Embrión no Mamífero , Femenino , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunosupresores/farmacología , Masculino , Morfolinos/farmacología , Proteínas de la Mielina/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
19.
Curr Biol ; 26(11): R468-70, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27269724

RESUMEN

The amount of myelin that forms on individual axons can vary considerably. Recent work, including a new study, indicates that myelin profiles on distinct subclasses of axons might be determined by diverse mechanisms.


Asunto(s)
Axones , Vaina de Mielina , Humanos
20.
Development ; 143(13): 2292-304, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226318

RESUMEN

The transition of dividing neuroepithelial progenitors to differentiated neurons and glia is essential for the formation of a functional nervous system. Sonic hedgehog (Shh) is a mitogen for spinal cord progenitors, but how cells become insensitive to the proliferative effects of Shh is not well understood. Because Shh reception occurs at primary cilia, which are positioned within the apical membrane of neuroepithelial progenitors, we hypothesized that loss of apical characteristics reduces the Shh signaling response, causing cell cycle exit and differentiation. We tested this hypothesis using genetic and pharmacological manipulation, gene expression analysis and time-lapse imaging of zebrafish embryos. Blocking the function of miR-219, a microRNA that downregulates apical Par polarity proteins and promotes progenitor differentiation, elevated Shh signaling. Inhibition of Shh signaling reversed the effects of miR-219 depletion and forced expression of Shh phenocopied miR-219 deficiency. Time-lapse imaging revealed that knockdown of miR-219 function accelerates the growth of primary cilia, revealing a possible mechanistic link between miR-219-mediated regulation of apical Par proteins and Shh signaling. Thus, miR-219 appears to decrease progenitor cell sensitivity to Shh signaling, thereby driving these cells towards differentiation.


Asunto(s)
Proteínas Hedgehog/metabolismo , MicroARNs/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Animales , Recuento de Células , Polaridad Celular , Cilios/metabolismo , Embrión no Mamífero/metabolismo , MicroARNs/genética , Mutación/genética , Organogénesis , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...