Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39265571

RESUMEN

Neurons have a unique polarized nature that must adapt to environmental changes throughout their lifespan. During embryonic development, axon elongation is led by the growth cone,1 culminating in the formation of a presynaptic terminal. After synapses are formed, axons elongate in a growth cone-independent manner to accompany body growth while maintaining their ultrastructure and function.2,3,4,5,6 To further understand mechanical strains on the axon shaft, we developed a computer-controlled stretchable microfluidic platform compatible with multi-omics and live imaging. Our data show that sensory embryonic dorsal root ganglia (DRGs) neurons have high plasticity, with axon shaft microtubules decreasing polymerization rates, aligning with the direction of tension, and undergoing stabilization. Moreover, in embryonic DRGs, stretch triggers yes-associated protein (YAP) nuclear translocation, supporting its participation in the regulatory network that enables tension-driven axon growth. Other than cytoskeleton remodeling, stretch prompted MARCKS-dependent formation of plasmalemmal precursor vesicles (PPVs), resulting in new membrane incorporation throughout the axon shaft. In contrast, adolescent DRGs showed a less robust adaptation, with axonal microtubules being less responsive to stretch. Also, while adolescent DRGs were still amenable to strain-induced PPV formation at higher stretch rates, new membrane incorporation in the axon shaft failed to occur. In summary, we developed a new resource to study the biology of axon stretch growth. By unraveling cytoskeleton adaptation and membrane remodeling in the axon shaft of stretched neurons, we are moving forward in understanding axon growth.

2.
Front Mol Neurosci ; 16: 1231659, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37588057

RESUMEN

Introduction: In Krabbe disease (KD), mutations in ß-galactosylceramidase (GALC), a lysosomal enzyme responsible for the catabolism of galactolipids, leads to the accumulation of its substrates galactocerebroside and psychosine. This neurologic condition is characterized by a severe and progressive demyelination together with neuron-autonomous defects and degeneration. Twitcher mice mimic the infantile form of KD, which is the most common form of the human disease. The Twitcher CNS and PNS present demyelination, axonal loss and neuronal defects including decreased levels of acetylated tubulin, decreased microtubule stability and impaired axonal transport. Methods: We tested whether inhibiting the α-tubulin deacetylase HDAC6 with a specific inhibitor, ACY-738, was able to counteract the early neuropathology and neuronal defects of Twitcher mice. Results: Our data show that delivery of ACY-738 corrects the low levels of acetylated tubulin in the Twitcher nervous system. Furthermore, it reverts the loss myelinated axons in the sciatic nerve and in the optic nerve when administered from birth to postnatal day 9, suggesting that the drug holds neuroprotective properties. The extended delivery of ACY-738 to Twitcher mice delayed axonal degeneration in the CNS and ameliorated the general presentation of the disease. ACY-738 was effective in rescuing neuronal defects of Twitcher neurons, stabilizing microtubule dynamics and increasing the axonal transport of mitochondria. Discussion: Overall, our results support that ACY-738 has a neuroprotective effect in KD and should be considered as an add-on therapy combined with strategies targeting metabolic correction.

3.
PLoS Biol ; 20(7): e3001706, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35793314

RESUMEN

In this issue of PLOS Biology, Kreher and colleagues show in a mouse model that in vivo, neurons and not only myelinating glia are primary effectors of disease progression in Krabbe disease. The neuron-specific model generated allows the unprecedented capacity to investigate the neuronal autonomous component of this disorder.


Asunto(s)
Galactosilceramidasa , Leucodistrofia de Células Globoides , Animales , Modelos Animales de Enfermedad , Galactosilceramidasa/genética , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/patología , Ratones , Neuroglía/patología , Neuronas/fisiología
4.
Brain ; 145(5): 1632-1640, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35661858

RESUMEN

The axon initial segment is a specialized compartment of the proximal axon of CNS neurons where action potentials are initiated. However, it remains unknown whether this domain is assembled in sensory dorsal root ganglion neurons, in which spikes are initiated in the peripheral terminals. Here we investigate whether sensory neurons have an axon initial segment and if it contributes to spontaneous activity in neuropathic pain. Our results demonstrate that myelinated dorsal root ganglion neurons assemble an axon initial segment in the proximal region of their stem axon, enriched in the voltage-gated sodium channels Nav1.1 and Nav1.7. Using correlative immunofluorescence and calcium imaging, we demonstrate that the Nav1.7 channels at the axon initial segment are associated with spontaneous activity. Computer simulations further indicate that the axon initial segment plays a key role in the initiation of spontaneous discharges by lowering their voltage threshold. Finally, using a Cre-based mouse model for time-controlled axon initial segment disassembly, we demonstrate that this compartment is a major source of spontaneous discharges causing mechanical allodynia in neuropathic pain. Thus, an axon initial segment domain is present in sensory neurons and facilitates their spontaneous activity. This study provides a new insight in the cellular mechanisms that cause pathological pain and identifies a new potential target for chronic pain management.


Asunto(s)
Segmento Inicial del Axón , Neuralgia , Animales , Ganglios Espinales/patología , Humanos , Hiperalgesia/patología , Ratones , Neuralgia/patología , Células Receptoras Sensoriales
5.
J Comp Neurol ; 530(12): 2215-2237, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35434782

RESUMEN

The African spiny mouse (Acomys cahirinus) is an emerging model of mammalian epimorphic regeneration that has aroused the interest of the scientific community in the last decade. To date, studies on brain repair have been hindered by the lack of knowledge on the neuroanatomy of this species. Here, we present a coronal brain atlas in stereotaxic coordinates, which allows for three-dimensional identification and localization of the brain structures of this species. The brain of 12-week-old spiny mice was mapped in stereotaxic coordinates using cresyl violet-stained brain sections obtained from coronal cryosectioning of the brain after transcardial perfusion with fixative. The atlas is presented in 42 plates representing sections spaced 240 µm apart. Stereotaxic coordinates were validated using both a model of Parkinsonian lesion of the striatum with 6-hydroxydopamine and labeling of the corticospinal tract in the spiny mouse spinal cord using AAV1/2-GFP intracortical injections. This work presents a new tool in A. cahirinus neurobiology and opens new avenues of research for the investigation of the regenerative ability of A. cahirinus in models of brain disorders.


Asunto(s)
Murinae , Médula Espinal , Animales , Encéfalo
6.
Dev Cell ; 57(4): 440-450.e7, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-34986324

RESUMEN

Regeneration of adult mammalian central nervous system (CNS) axons is abortive, resulting in inability to recover function after CNS lesion, including spinal cord injury (SCI). Here, we show that the spiny mouse (Acomys) is an exception to other mammals, being capable of spontaneous and fast restoration of function after severe SCI, re-establishing hind limb coordination. Remarkably, Acomys assembles a scarless pro-regenerative tissue at the injury site, providing a unique structural continuity of the initial spinal cord geometry. The Acomys SCI site shows robust axon regeneration of multiple tracts, synapse formation, and electrophysiological signal propagation. Transcriptomic analysis of the spinal cord following transcriptome reconstruction revealed that Acomys rewires glycosylation biosynthetic pathways, culminating in a specific pro-regenerative proteoglycan signature at SCI site. Our work uncovers that a glycosylation switch is critical for axon regeneration after SCI and identifies ß3gnt7, a crucial enzyme of keratan sulfate biosynthesis, as an enhancer of axon growth.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/patología , Animales , Axones/patología , Modelos Animales de Enfermedad , Glicosilación , Ratones , Médula Espinal/fisiología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología , Columna Vertebral/fisiopatología
7.
Genet Med ; 24(2): 319-331, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906466

RESUMEN

PURPOSE: Adducins interconnect spectrin and actin filaments to form polygonal scaffolds beneath the cell membranes and form ring-like structures in neuronal axons. Adducins regulate mouse neural development, but their function in the human brain is unknown. METHODS: We used exome sequencing to uncover ADD1 variants associated with intellectual disability (ID) and brain malformations. We studied ADD1 splice isoforms in mouse and human neocortex development with RNA sequencing, super resolution imaging, and immunoblotting. We investigated 4 variant ADD1 proteins and heterozygous ADD1 cells for protein expression and ADD1-ADD2 dimerization. We studied Add1 functions in vivo using Add1 knockout mice. RESULTS: We uncovered loss-of-function ADD1 variants in 4 unrelated individuals affected by ID and/or structural brain defects. Three additional de novo copy number variations covering the ADD1 locus were associated with ID and brain malformations. ADD1 is highly expressed in the neocortex and the corpus callosum, whereas ADD1 splice isoforms are dynamically expressed between cortical progenitors and postmitotic neurons. Human variants impair ADD1 protein expression and/or dimerization with ADD2. Add1 knockout mice recapitulate corpus callosum dysgenesis and ventriculomegaly phenotypes. CONCLUSION: Our human and mouse genetics results indicate that pathogenic ADD1 variants cause corpus callosum dysgenesis, ventriculomegaly, and/or ID.


Asunto(s)
Hidrocefalia , Discapacidad Intelectual , Agenesia del Cuerpo Calloso/genética , Agenesia del Cuerpo Calloso/patología , Animales , Variaciones en el Número de Copia de ADN , Humanos , Hidrocefalia/genética , Discapacidad Intelectual/genética , Ratones , Fenotipo
8.
Front Cell Dev Biol ; 9: 747699, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34820375

RESUMEN

Transthyretin (TTR), a plasma and cerebrospinal fluid protein, increases axon growth and organelle transport in sensory neurons. While neurons extend their axons, the microtubule (MT) cytoskeleton is crucial for the segregation of functional compartments and axonal outgrowth. Herein, we investigated whether TTR promotes axon elongation by modulating MT dynamics. We found that TTR KO mice have an intrinsic increase in dynamic MTs and reduced levels of acetylated α-tubulin in peripheral axons. In addition, they failed to modulate MT dynamics in response to sciatic nerve injury, leading to decreased regenerative capacity. Importantly, restoring acetylated α-tubulin levels of TTR KO dorsal root ganglia (DRG) neurons using an HDAC6 inhibitor is sufficient to completely revert defective MT dynamics and neurite outgrowth. In summary, our results reveal a new role for TTR in the modulation of MT dynamics by regulating α-tubulin acetylation via modulation of the acetylase ATAT1, and suggest that this activity underlies TTR neuritogenic function.

9.
Dev Neurobiol ; 81(3): 300-309, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32302060

RESUMEN

Throughout development, neurons are capable of integrating external and internal signals leading to the morphological changes required for neuronal polarization and axon growth. The first phase of axon elongation occurs during neuronal polarization. At this stage, membrane remodeling and cytoskeleton dynamics are crucial for the growth cone to advance and guide axon elongation. When a target is recognized, the growth cone collapses to form the presynaptic terminal. Once a synapse is established, the growth of the organism results in an increased distance between the neuronal cell bodies and their targets. In this second phase of axon elongation, growth cone-independent molecular mechanisms and cytoskeleton changes must occur to enable axon growth to accompany the increase in body size. While the field has mainly focused on growth-cone mediated axon elongation during development, tension driven axon growth remains largely unexplored. In this review, we will discuss in a critical perspective the current knowledge on the mechanisms guiding axon growth following synaptogenesis, with a particular focus on the putative role played by the axonal cytoskeleton.


Asunto(s)
Axones , Citoesqueleto , Axones/fisiología , Conos de Crecimiento , Microtúbulos/fisiología , Neuronas/fisiología
10.
Cells ; 9(9)2020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32858875

RESUMEN

By binding to actin filaments, non-muscle myosin II (NMII) generates actomyosin networks that hold unique contractile properties. Their dynamic nature is essential for neuronal biology including the establishment of polarity, growth cone formation and motility, axon growth during development (and axon regeneration in the adult), radial and longitudinal axonal tension, and synapse formation and function. In this review, we discuss the current knowledge on the spatial distribution and function of the actomyosin cytoskeleton in different axonal compartments. We highlight some of the apparent contradictions and open questions in the field, including the role of NMII in the regulation of axon growth and regeneration, the possibility that NMII structural arrangement along the axon shaft may control both radial and longitudinal contractility, and the mechanism and functional purpose underlying NMII enrichment in the axon initial segment. With the advances in live cell imaging and super resolution microscopy, it is expected that in the near future the spatial distribution of NMII in the axon, and the mechanisms by which it participates in axonal biology will be further untangled.


Asunto(s)
Axones/metabolismo , Conos de Crecimiento/metabolismo , Humanos
11.
Elife ; 92020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32195665

RESUMEN

Neurons have a membrane periodic skeleton (MPS) composed of actin rings interconnected by spectrin. Here, combining chemical and genetic gain- and loss-of-function assays, we show that in rat hippocampal neurons the MPS is an actomyosin network that controls axonal expansion and contraction. Using super-resolution microscopy, we analyzed the localization of axonal non-muscle myosin II (NMII). We show that active NMII light chains are colocalized with actin rings and organized in a circular periodic manner throughout the axon shaft. In contrast, NMII heavy chains are mostly positioned along the longitudinal axonal axis, being able to crosslink adjacent rings. NMII filaments can play contractile or scaffolding roles determined by their position relative to actin rings and activation state. We also show that MPS destabilization through NMII inactivation affects axonal electrophysiology, increasing action potential conduction velocity. In summary, our findings open new perspectives on axon diameter regulation, with important implications in neuronal biology.


Asunto(s)
Actomiosina/fisiología , Axones/fisiología , Conducción Nerviosa/fisiología , Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Animales , Línea Celular , Humanos , Ratones , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIB no Muscular/genética , Ratas
13.
J Clin Invest ; 130(4): 2024-2040, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31945017

RESUMEN

After trauma, regeneration of adult CNS axons is abortive, causing devastating neurologic deficits. Despite progress in rehabilitative care, there is no effective treatment that stimulates axonal growth following injury. Using models with different regenerative capacities, followed by gain- and loss-of-function analysis, we identified profilin 1 (Pfn1) as a coordinator of actin and microtubules (MTs), powering axonal growth and regeneration. In growth cones, Pfn1 increased actin retrograde flow, MT growth speed, and invasion of filopodia by MTs, orchestrating cytoskeletal dynamics toward axonal growth. In vitro, active Pfn1 promoted MT growth in a formin-dependent manner, whereas localization of MTs to growth cone filopodia was facilitated by direct MT binding and interaction with formins. In vivo, Pfn1 ablation limited regeneration of growth-competent axons after sciatic nerve and spinal cord injury. Adeno-associated viral (AAV) delivery of constitutively active Pfn1 to rodents promoted axonal regeneration, neuromuscular junction maturation, and functional recovery of injured sciatic nerves, and increased the ability of regenerating axons to penetrate the inhibitory spinal cord glial scar. Thus, we identify Pfn1 as an important regulator of axonal regeneration and suggest that AAV-mediated delivery of constitutively active Pfn1, together with the identification of modulators of Pfn1 activity, should be considered to treat the injured nervous system.


Asunto(s)
Citoesqueleto , Terapia Genética , Conos de Crecimiento/metabolismo , Regeneración Nerviosa , Nervio Ciático/fisiología , Traumatismos de la Médula Espinal , Animales , Citoesqueleto/genética , Citoesqueleto/metabolismo , Dependovirus , Ratones , Ratones Noqueados , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Profilinas/biosíntesis , Profilinas/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia , Transducción Genética
14.
Cytoskeleton (Hoboken) ; 77(3-4): 76-83, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31811707

RESUMEN

Although originally identified as G-actin sequestering proteins, profilins are emerging as critical regulators of actin dynamics, capable of interacting with multiple acting binding proteins, and being able to link membrane lipids to cytoskeleton components. Recently, in addition to its actin, poly-proline, and phosphatidylinositol binding domains, profilin has been shown to contain residues specialized in microtubule binding. Here we will discuss in a critical perspective the emerging body of data supporting that profilins are central mediators of actin microfilament and microtubule interaction. We will also address the unanswered questions in the field, including the nature of the interaction of profilin with microtubules, and its effect on microtubule dynamics. These recent discoveries deepen our understanding on how different cytoskeleton components are integrated within cells.


Asunto(s)
Actinas/metabolismo , Microtúbulos/metabolismo , Profilinas/metabolismo , Unión Proteica/fisiología , Humanos
15.
Neurourol Urodyn ; 38(6): 1540-1550, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31180583

RESUMEN

OBJECTIVES: To investigate if intravesical administration during spinal shock of resiniferatoxin (RTX), an ultrapotent desensitizing agonist of transient receptor potential vanilloid-1 (TRPV1), would silence TRPV1-expressing bladder afferents at an early stage of disease progression and modulate neurogenic detrusor overactivity (NDO) emergence. MATERIALS AND METHODS: Rats submitted to largely incomplete spinal cord transection at T8/9 spinal segment were treated with intravesical RTX (50 nM) or its vehicle during spinal shock. Four weeks after spinal lesion, bladder-reflex activity was evaluated by cystometry under urethane anesthesia, after which the bladder, spinal cord, and dorsal root ganglia were collected and processed. RESULTS: We found improvements on bladder function several weeks after early intravesical RTX administration, including a marked decrease of intravesical pressures and amplitude of bladder contractions. Such strong long-lasting urodynamic effects resulted from the very potent desensitizing activity of RTX on peripheral terminals of sensory afferents, an effect restricted to the bladder. CONCLUSION: Our results support that an early intervention with RTX could potentially attenuate NDO development and ensuing urinary incontinence, with a dramatic impact on the quality of life of spinal cord injury patients.


Asunto(s)
Diterpenos/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Vejiga Urinaria Hiperactiva/etiología , Administración Intravesical , Animales , Péptido Relacionado con Gen de Calcitonina/biosíntesis , Diterpenos/administración & dosificación , Femenino , Proteína GAP-43/biosíntesis , Ganglios Espinales/diagnóstico por imagen , Neuronas Aferentes , Ratas , Ratas Wistar , Reflejo , Traumatismos de la Médula Espinal/fisiopatología , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/biosíntesis , Vejiga Urinaria/inervación , Vejiga Urinaria/fisiopatología , Urodinámica/efectos de los fármacos
16.
Mol Ther Nucleic Acids ; 11: 393-406, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29858074

RESUMEN

After spinal cord injury (SCI), nerve regeneration is severely hampered due to the establishment of a highly inhibitory microenvironment at the injury site, through the contribution of multiple factors. The potential of antisense oligonucleotides (AONs) to modify gene expression at different levels, allowing the regulation of cell survival and cell function, together with the availability of chemically modified nucleic acids with favorable biopharmaceutical properties, make AONs an attractive tool for novel SCI therapy developments. In this work, we explored the potential of locked nucleic acid (LNA)-modified AON gapmers in combination with a fibrin hydrogel bridging material to induce gene silencing in situ at a SCI lesion site. LNA gapmers were effectively developed against two promising gene targets aiming at enhancing axonal regeneration-RhoA and GSK3ß. The fibrin-matrix-assisted AON delivery system mediated potent RNA knockdown in vitro in a dorsal root ganglion explant culture system and in vivo at a SCI lesion site, achieving around 75% downregulation 5 days after hydrogel injection. Our results show that local implantation of a AON-gapmer-loaded hydrogel matrix mediated efficient gene silencing in the lesioned spinal cord and is an innovative platform that can potentially combine gene regulation with regenerative permissive substrates aiming at SCI therapeutics and nerve regeneration.

17.
Neurobiol Aging ; 59: 10-14, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28780366

RESUMEN

The deposition of amyloid ß peptide (Aß) in the hippocampus is one of the major hallmarks of Alzheimer's disease, a neurodegenerative disorder characterized by memory loss and cognitive impairment. The modulation of Aß levels in the brain results from an equilibrium between its production from the amyloid precursor protein and removal by amyloid clearance proteins, which might occur via enzymatic (Aß-degrading enzymes) or nonenzymatic (binding/transport proteins) reactions. Transthyretin (TTR) is one of the major Aß-binding proteins acting as a neuroprotector in AD. In addition, TTR cleaves Aß peptide in vitro. In this work, we show that proteolytically active TTR, and not the inactive form of the protein, impacts on Aß fibrillogenesis, degrades neuronal-secreted Aß, and reduces Aß-induced toxicity in hippocampal neurons. Our data demonstrate that TTR proteolytic activity is required for the neuroprotective effect of the protein constituting a putative novel therapeutic target for AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Fármacos Neuroprotectores , Prealbúmina/fisiología , Proteolisis , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Células Cultivadas , Humanos , Terapia Molecular Dirigida , Prealbúmina/genética , Prealbúmina/metabolismo , Unión Proteica
18.
Acta Biomater ; 59: 243-256, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694236

RESUMEN

To enhance fibrin hydrogel affinity towards pluripotent stem cell-derived neural stem/progenitor cells (NSPCs) and its capacity to support NSPC migration and neurite extension, we explored the tethering of synthetic peptides engaging integrin α6ß1, a cell receptor enriched in NSPCs. Six α6ß1 integrin ligands were tested for their ability to support integrin α6ß1-mediated adhesion of embryonic stem cell-derived NSPCs (ES-NSPs) and sustain ES-NSPC viability, migration, and neuronal differentiation. Due to their better performance, peptides T1, HYD1, and A5G81 were immobilized into fibrin and functionalized gels characterized in terms of peptide binding efficiency, structure and viscoelastic properties. Tethering of T1 or HYD1 successfully enhanced cell outgrowth from ES-NSPC neurospheres (up to 2.4-fold increase), which exhibited a biphasic response to peptide concentration. Inhibition assays evidenced the involvement of α6ß1 and α3ß1 integrins in mediating radial outgrowth on T1-/HYD1-functionalized gels. Fibrin functionalization also promoted neurite extension of single ES-NSPCs in fibrin, without affecting cell proliferation and neuronal differentiation. Finally, HYD1-functionalized gels were found to provide a permissive environment for axonal regeneration, leading up to a 2.0-fold increase in neurite extension from rat dorsal root ganglia explants as compared to unmodified fibrin, and to significant improved locomotor function after spinal cord injury (complete transection), along with a trend toward a higher area positive for growth associated protein 43 (marker for axonal growth cone formation). Our results suggest that conjugation of α6ß1 integrin-binding motifs is of interest to increase the biofunctionality of hydrogels used in 3D platforms for ES-NSPC culture and potentially, in matrix-assisted ES-NSPC transplantation. STATEMENT OF SIGNIFICANCE: Impact statement: The transplantation of NSPCs derived from pluripotent stem cells holds much promise for the treatment of central nervous system disorders. Moreover, the combinatorial use of biodegradable hydrogels with NSPCs was shown to contribute to the establishment of a more permissive environment for survival and integration of transplanted cells. In this study, fibrin hydrogels functionalized with a synthetic peptide engaging integrin α6ß1 (HYD1) were shown to promote neurite extension of ES-NSPCs, which is fundamental for the formation of functional neuronal relay circuits after NSPC transplantation. Notably, HYD1-functionalized fibrin per se led to enhanced axonal growth ex vivo and to an improvement in locomotor function after implantation in a rat model of spinal cord injury. Conjugation of α6ß1 integrin-binding motifs may therefore be of interest to confer bioactivity to NSPC hydrogel vehicles.


Asunto(s)
Células Madre Embrionarias/metabolismo , Fibrina/química , Integrina alfa6beta1/metabolismo , Células-Madre Neurales/metabolismo , Neuritas/metabolismo , Animales , Línea Celular Tumoral , Células Madre Embrionarias/citología , Humanos , Ligandos , Ratones , Células-Madre Neurales/citología , Ratas , Ratas Wistar
19.
Cereb Cortex ; 27(3): 1732-1747, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334068

RESUMEN

KIAA0319 is a transmembrane protein associated with dyslexia with a presumed role in neuronal migration. Here we show that KIAA0319 expression is not restricted to the brain but also occurs in sensory and spinal cord neurons, increasing from early postnatal stages to adulthood and being downregulated by injury. This suggested that KIAA0319 participates in functions unrelated to neuronal migration. Supporting this hypothesis, overexpression of KIAA0319 repressed axon growth in hippocampal and dorsal root ganglia neurons; the intracellular domain of KIAA0319 was sufficient to elicit this effect. A similar inhibitory effect was observed in vivo as axon regeneration was impaired after transduction of sensory neurons with KIAA0319. Conversely, the deletion of Kiaa0319 in neurons increased neurite outgrowth in vitro and improved axon regeneration in vivo. At the mechanistic level, KIAA0319 engaged the JAK2-SH2B1 pathway to activate Smad2, which played a central role in KIAA0319-mediated repression of axon growth. In summary, we establish KIAA0319 as a novel player in axon growth and regeneration with the ability to repress the intrinsic growth potential of axons. This study describes a novel regulatory mechanism operating during peripheral nervous system and central nervous system axon growth, and offers novel targets for the development of effective therapies to promote axon regeneration.


Asunto(s)
Axones/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proyección Neuronal , Proteína Smad2/metabolismo , Envejecimiento/metabolismo , Animales , Aumento de la Célula , Línea Celular , Células Cultivadas , Femenino , Ganglios Espinales/metabolismo , Hipocampo/metabolismo , Humanos , Janus Quinasa 2/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Dominios Proteicos , Ratas Wistar , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Médula Espinal/metabolismo
20.
Brain Struct Funct ; 221(5): 2777-99, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26068054

RESUMEN

Axon growth and dendrite development are key processes for the establishment of a functional neuronal network. Adenosine, which is released by neurons and glia, is a known modulator of synaptic transmission but its influence over neuronal growth has been much less investigated. We now explored the action of adenosine A2A receptors (A2AR) upon neurite outgrowth, discriminating actions over the axon or dendrites, and the mechanisms involved. Morphometric analysis of primary cultures of cortical neurons from E18 Sprague-Dawley rats demonstrated that an A2AR agonist, CGS 21680, enhances axonal elongation and dendritic branching, being the former prevented by inhibitors of phosphoinositide 3-kinase, mitogen-activated protein kinase and phospholipase C, but not of protein kinase A. By testing the influence of a scavenger of BDNF (brain-derived neurotrophic factor) over the action of the A2AR agonist and the action of a selective A2AR antagonist over the action of BDNF, we could conclude that while the action of A2ARs upon dendritic branching is dependent on the presence of endogenous BDNF, the influence of A2ARs upon axonal elongation is independent of endogenous BDNF. In consonance with the action over axonal elongation, A2AR activation promoted a decrease in microtubule stability and an increase in microtubule growth speed in axonal growth cones. In conclusion, we disclose a facilitatory action of A2ARs upon axonal elongation and microtubule dynamics, providing new insights for A2ARs regulation of neuronal differentiation and axonal regeneration.


Asunto(s)
Axones/fisiología , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Dendritas/fisiología , Neuronas/fisiología , Receptor de Adenosina A2A/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Animales , Axones/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Dendritas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/fisiología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Fenetilaminas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor trkB/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA