Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 24(6): 396-413, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36604586

RESUMEN

One hundred years ago, Ramón y Cajal, considered by many as the founder of modern neuroscience, stated that neurons of the adult central nervous system (CNS) are incapable of regenerating. Yet, recent years have seen a tremendous expansion of knowledge in the molecular control of axon regeneration after CNS injury. We now understand that regeneration in the adult CNS is limited by (1) a failure to form cellular or molecular substrates for axon attachment and elongation through the lesion site; (2) environmental factors, including inhibitors of axon growth associated with myelin and the extracellular matrix; (3) astrocyte responses, which can both limit and support axon growth; and (4) intraneuronal mechanisms controlling the establishment of an active cellular growth programme. We discuss these topics together with newly emerging hypotheses, including the surprising finding from transcriptomic analyses of the corticospinal system in mice that neurons revert to an embryonic state after spinal cord injury, which can be sustained to promote regeneration with neural stem cell transplantation. These gains in knowledge are steadily advancing efforts to develop effective treatment strategies for spinal cord injury in humans.


Asunto(s)
Axones , Traumatismos de la Médula Espinal , Humanos , Ratones , Animales , Axones/patología , Axones/fisiología , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/terapia , Traumatismos de la Médula Espinal/patología , Neuronas/fisiología , Mamíferos
2.
Annu Rev Cell Dev Biol ; 35: 501-521, 2019 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-31590586

RESUMEN

The dual leucine zipper-bearing kinase (DLK) and leucine zipper-bearing kinase (LZK) are evolutionarily conserved MAPKKKs of the mixed-lineage kinase family. Acting upstream of stress-responsive JNK and p38 MAP kinases, DLK and LZK have emerged as central players in neuronal responses to a variety of acute and traumatic injuries. Recent studies also implicate their function in astrocytes, microglia, and other nonneuronal cells, reflecting their expanding roles in the multicellular response to injury and in disease. Of particular note is the potential link of these kinases to neurodegenerative diseases and cancer. It is thus critical to understand the physiological contexts under which these kinases are activated, as well as the signal transduction mechanisms that mediate specific functional outcomes. In this review we first provide a historical overview of the biochemical and functional dissection of these kinases. We then discuss recent findings on regulating their activity to enhance cellular protection following injury and in disease, focusing on but not limited to the nervous system.


Asunto(s)
Leucina Zippers/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Neuronas/metabolismo , Estrés Fisiológico/genética , Animales , Axones/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/virología , Neuroglía/metabolismo , Neuronas/virología , Regeneración/genética , Regeneración/fisiología , Células Madre/metabolismo , Estrés Fisiológico/fisiología , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
3.
Cell ; 150(6): 1264-73, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22980985

RESUMEN

Neural stem cells (NSCs) expressing GFP were embedded into fibrin matrices containing growth factor cocktails and grafted to sites of severe spinal cord injury. Grafted cells differentiated into multiple cellular phenotypes, including neurons, which extended large numbers of axons over remarkable distances. Extending axons formed abundant synapses with host cells. Axonal growth was partially dependent on mammalian target of rapamycin (mTOR), but not Nogo signaling. Grafted neurons supported formation of electrophysiological relays across sites of complete spinal transection, resulting in functional recovery. Two human stem cell lines (566RSC and HUES7) embedded in growth-factor-containing fibrin exhibited similar growth, and 566RSC cells supported functional recovery. Thus, properties intrinsic to early-stage neurons can overcome the inhibitory milieu of the injured adult spinal cord to mount remarkable axonal growth, resulting in formation of new relay circuits that significantly improve function. These therapeutic properties extend across stem cell sources and species.


Asunto(s)
Axones/fisiología , Células-Madre Neurales/trasplante , Traumatismos de la Médula Espinal/terapia , Regeneración de la Medula Espinal , Animales , Línea Celular , Femenino , Proteínas Fluorescentes Verdes/análisis , Humanos , Células-Madre Neurales/citología , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Médula Espinal/patología , Médula Espinal/fisiopatología
4.
Nature ; 581(7806): 77-82, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32376949

RESUMEN

Grafts of spinal-cord-derived neural progenitor cells (NPCs) enable the robust regeneration of corticospinal axons and restore forelimb function after spinal cord injury1; however, the molecular mechanisms that underlie this regeneration are unknown. Here we perform translational profiling specifically of corticospinal tract (CST) motor neurons in mice, to identify their 'regenerative transcriptome' after spinal cord injury and NPC grafting. Notably, both injury alone and injury combined with NPC grafts elicit virtually identical early transcriptomic responses in host CST neurons. However, in mice with injury alone this regenerative transcriptome is downregulated after two weeks, whereas in NPC-grafted mice this transcriptome is sustained. The regenerative transcriptome represents a reversion to an embryonic transcriptional state of the CST neuron. The huntingtin gene (Htt) is a central hub in the regeneration transcriptome; deletion of Htt significantly attenuates regeneration, which shows that Htt has a key role in neural plasticity after injury.


Asunto(s)
Proliferación Celular/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Regeneración Nerviosa/genética , Células-Madre Neurales/citología , Neuronas/metabolismo , Neuronas/patología , Transcripción Genética , Animales , Axones/patología , Axones/fisiología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Proteína Huntingtina/genética , Ratones , Células-Madre Neurales/trasplante , Plasticidad Neuronal , Neuronas/citología , Neuronas/trasplante , Biosíntesis de Proteínas , Tractos Piramidales/citología , Tractos Piramidales/metabolismo , Tractos Piramidales/patología , RNA-Seq , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Transcriptoma
6.
J Neurosci ; 42(18): 3716-3732, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35361703

RESUMEN

The limited ability for axonal repair after spinal cord injury underlies long-term functional impairment. Dual leucine-zipper kinase [DLK; MAP kinase kinase kinase 12; MAP3K12] is an evolutionarily conserved MAP3K implicated in neuronal injury signaling from Caenorhabditis elegans to mammals. However, whether DLK or its close homolog leucine zipper kinase (LZK; MAP3K13) regulates axonal repair in the mammalian spinal cord remains unknown. Here, we assess the role of endogenous DLK and LZK in the regeneration and compensatory sprouting of corticospinal tract (CST) axons in mice of both sexes with genetic analyses in a regeneration competent background provided by PTEN deletion. We found that inducible neuronal deletion of both DLK and LZK, but not either kinase alone, abolishes PTEN deletion-induced regeneration and sprouting of CST axons, and reduces naturally-occurring axon sprouting after injury. Thus, DLK/LZK-mediated injury signaling operates not only in injured neurons to regulate regeneration, but also unexpectedly in uninjured neurons to regulate sprouting. Deleting DLK and LZK does not interfere with PTEN/mTOR signaling, indicating that injury signaling and regenerative competence are independently controlled. Together with our previous study implicating LZK in astrocytic reactivity and scar formation, these data illustrate the multicellular function of this pair of MAP3Ks in both neurons and glia in the injury response of the mammalian spinal cord.SIGNIFICANCE STATEMENT Functional recovery after spinal cord injury is limited because of a lack of axonal repair in the mammalian CNS. Dual leucine-zipper kinase (DLK) and leucine zipper kinase (LZK) are two closely related protein kinases that have emerged as regulators of neuronal responses to injury. However, their role in axonal repair in the mammalian spinal cord has not been described. Here, we show that DLK and LZK together play critical roles in axonal repair in the mammalian spinal cord, validating them as potential targets to promote repair and recovery after spinal cord injury. In addition to regulating axonal regeneration from injured neurons, both kinases also regulate compensatory axonal growth from uninjured neurons, indicating a more pervasive role in CNS repair than originally anticipated.


Asunto(s)
Leucina Zippers , Quinasas Quinasa Quinasa PAM/metabolismo , Traumatismos de la Médula Espinal , Animales , Axones/fisiología , Femenino , Leucina/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Masculino , Mamíferos , Ratones , Regeneración Nerviosa/fisiología , Tractos Piramidales/fisiología
7.
Glia ; 71(10): 2473-2494, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37401784

RESUMEN

Nogo-A, B, and C are well described members of the reticulon family of proteins, most well known for their negative regulatory effects on central nervous system (CNS) neurite outgrowth and repair following injury. Recent research indicates a relationship between Nogo-proteins and inflammation. Microglia, the brain's immune cells and inflammation-competent compartment, express Nogo protein, although specific roles of the Nogo in these cells is understudied. To examine inflammation-related effects of Nogo, we generated a microglial-specific inducible Nogo KO (MinoKO) mouse and challenged the mouse with a controlled cortical impact (CCI) traumatic brain injury (TBI). Histological analysis shows no difference in brain lesion sizes between MinoKO-CCI and Control-CCI mice, although MinoKO-CCI mice do not exhibit the levels of ipsilateral lateral ventricle enlargement as injury matched controls. Microglial Nogo-KO results in decreased lateral ventricle enlargement, microglial and astrocyte immunoreactivity, and increased microglial morphological complexity compared to injury matched controls, suggesting decreased tissue inflammation. Behaviorally, healthy MinoKO mice do not differ from control mice, but automated tracking of movement around the home cage and stereotypic behavior, such as grooming and eating (termed cage "activation"), following CCI is significantly elevated. Asymmetrical motor function, a deficit typical of unilaterally brain lesioned rodents, was not detected in CCI injured MinoKO mice, while the phenomenon was present in CCI injured controls 1-week post-injury. Overall, our studies show microglial Nogo as a negative regulator of recovery following brain injury. To date, this is the first evaluation of the roles microglial specific Nogo in a rodent injury model.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Proteínas Nogo , Animales , Ratones , Lesiones Encefálicas/patología , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Proteínas Nogo/metabolismo
8.
J Neurosci ; 37(45): 10808-10816, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118209

RESUMEN

In vivo optical imaging has emerged as a powerful tool with which to study cellular responses to injury and disease in the mammalian CNS. Important new insights have emerged regarding axonal degeneration and regeneration, glial responses and neuroinflammation, changes in the neurovascular unit, and, more recently, neural transplantations. Accompanying a 2017 SfN Mini-Symposium, here, we discuss selected recent advances in understanding the neuronal, glial, and other cellular responses to CNS injury and disease with in vivo imaging of the rodent brain or spinal cord. We anticipate that in vivo optical imaging will continue to be at the forefront of breakthrough discoveries of fundamental mechanisms and therapies for CNS injury and disease.


Asunto(s)
Enfermedades del Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/lesiones , Neuroimagen/métodos , Animales , Humanos , Ratones , Neuroimagen/instrumentación , Ratas
9.
J Neurosci ; 35(16): 6413-28, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904793

RESUMEN

Axons in the adult CNS have poor ability to grow after injury, impeding functional recovery in patients of spinal cord injury. This has been attributed to both a developmental decline in neuron-intrinsic growth ability and the presence of extrinsic growth inhibitors. We previously showed that genetic deletion of Nogo, an extrinsic inhibitor, promoted axonal sprouting from uninjured corticospinal tract (CST) neurons but not regeneration from injured CST neurons, whereas genetic deletion of PTEN, an intrinsic inhibitor, promoted both CST sprouting and regeneration. Here we test the hypothesis that combining an elevation of neuron-intrinsic growth ability and a reduction of extrinsic growth inhibition by genetic codeletion of PTEN and Nogo may further improve injury-induced axonal growth. In an apparent paradox, additionally deleting Nogo further enhanced CST regeneration but not sprouting in PTEN-deleted mice. Enhanced CST regeneration and sprouting in PTEN and PTEN/Nogo-deleted mice were associated with no or only temporary improvement in functional recovery. Our data illustrate that neuron-intrinsic and -extrinsic factors regulate axon regeneration and sprouting in complex ways and provide proof-of-principle evidence that targeting both can further improve regeneration. Neuron-intrinsic growth ability is an important determinant of neuronal responsiveness to changes in extrinsic growth inhibition, such that an elevated intrinsic growth state is a prerequisite for reducing extrinsic inhibition to take effect on CST regeneration. Meanwhile, additional strategies are required to unleash the full potential for functional recovery with enhanced axon regeneration and/or sprouting.


Asunto(s)
Axones/fisiología , Proteínas de la Mielina/deficiencia , Regeneración Nerviosa/fisiología , Fosfohidrolasa PTEN/deficiencia , Tractos Piramidales/fisiología , Animales , Conducta Animal/fisiología , Ratones , Ratones Mutantes , Proteínas de la Mielina/genética , Proteínas de la Mielina/fisiología , Regeneración Nerviosa/genética , Proteínas Nogo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Recuperación de la Función/genética , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología
10.
Proc Natl Acad Sci U S A ; 109(4): 1299-304, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22160722

RESUMEN

A requisite component of nervous system development is the achievement of cellular recognition and spatial segregation through competition-based refinement mechanisms. Competition for available axon space by myelinating oligodendrocytes ensures that all relevant CNS axons are myelinated properly. To ascertain the nature of this competition, we generated a transgenic mouse with sparsely labeled oligodendrocytes and establish that individual oligodendrocytes occupying similar axon tracts can greatly vary the number and lengths of their myelin internodes. Here we show that intercellular interactions between competing oligodendroglia influence the number and length of myelin internodes, referred to as myelinogenic potential, and identify the amino-terminal region of Nogo-A, expressed by oligodendroglia, as necessary and sufficient to inhibit this process. Exuberant and expansive myelination/remyelination is detected in the absence of Nogo during development and after demyelination, suggesting that spatial segregation and myelin extent is limited by microenvironmental inhibition. We demonstrate a unique physiological role for Nogo-A in the precise myelination of the developing CNS. Maximizing the myelinogenic potential of oligodendrocytes may offer an effective strategy for repair in future therapies for demyelination.


Asunto(s)
Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/fisiopatología , Proteínas de la Mielina/metabolismo , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Animales , Western Blotting , Sistema Nervioso Central/citología , Técnicas de Silenciamiento del Gen , Técnicas Histológicas , Ratones , Ratones Transgénicos , Microscopía Electrónica , Microesferas , Proteínas de la Mielina/genética , Proteínas Nogo , Oligodendroglía/metabolismo , Oligodendroglía/ultraestructura , Poliestirenos , ARN Interferente Pequeño/genética , Ultracentrifugación
11.
J Neurosci ; 32(15): 5209-15, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22496566

RESUMEN

Repulsive Eph forward signaling from limb-derived ephrins guides the axons of lateral motor column (LMC) motor neurons. LMC axons also express ephrinAs, while their EphA receptors are expressed in the limb mesenchyme. In vitro studies have suggested that reverse signaling from limb-derived EphA4 to axonal ephrinAs might result in attraction of LMC axons. However, genetic evidence for this function is lacking. Here we use the Dunn chamber turning assay to show that EphA proteins are chemoattractants and elicit fast turning responses in LMC neurons in vitro. Moreover, ectopic expression of EphA4 in chick hindlimb changes the limb trajectory of LMC axons. Nervous system-specific deletion of EphA4 in mice resulted in fewer LMC axon projection errors than the ubiquitous deletion of EphA4. Additionally, a signaling-incompetent EphA4 mutant partially rescued guidance errors in the hindlimb, suggesting that limb-derived EphA4 contributes to the establishment of LMC projections. In summary, we provide evidence for a role of EphA:ephrinA attractive reverse signaling in motor axon guidance and in vivo evidence of in-parallel forward Eph and reverse ephrin signaling function in the same neuronal population.


Asunto(s)
Axones/fisiología , Movimiento Celular/fisiología , Efrinas/genética , Efrinas/fisiología , Neuronas Motoras/fisiología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Embrión de Pollo , Vías Eferentes/citología , Vías Eferentes/fisiología , Electroforesis en Gel de Poliacrilamida , Miembro Posterior/inervación , Miembro Posterior/fisiología , Inmunohistoquímica , Ratones , Ratones Noqueados , Receptores de la Familia Eph/metabolismo
12.
Res Sq ; 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36865182

RESUMEN

The corticospinal tract (CST) is clinically important for the recovery of motor functions after spinal cord injury. Despite substantial progress in understanding the biology of axon regeneration in the central nervous system (CNS), our ability to promote CST regeneration remains limited. Even with molecular interventions, only a small proportion of CST axons regenerate1. Here we investigate this heterogeneity in the regenerative ability of corticospinal neurons following PTEN and SOCS3 deletion with patch-based single cell RNA sequencing (scRNA-Seq)2,3, which enables deep sequencing of rare regenerating neurons. Bioinformatic analyses highlighted the importance of antioxidant response and mitochondrial biogenesis along with protein translation. Conditional gene deletion validated a role for NFE2L2 (or NRF2), a master regulator of antioxidant response, in CST regeneration. Applying Garnett4, a supervised classification method, to our dataset gave rise to a Regenerating Classifier (RC), which, when applied to published scRNA-Seq data, generates cell type- and developmental stage-appropriate classifications. While embryonic brain, adult dorsal root ganglion and serotonergic neurons are classified as Regenerators, most neurons from adult brain and spinal cord are classified as Non-regenerators. Adult CNS neurons partially revert to a regenerative state soon after injury, which is accelerated by molecular interventions. Our data indicate the existence of universal transcriptomic signatures underlying the regenerative abilities of vastly different neuronal populations, and further illustrate that deep sequencing of only hundreds of phenotypically identified CST neurons has the power to reveal new insights into their regenerative biology.

13.
Neuron ; 111(24): 3953-3969.e5, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37848024

RESUMEN

Despite substantial progress in understanding the biology of axon regeneration in the CNS, our ability to promote regeneration of the clinically important corticospinal tract (CST) after spinal cord injury remains limited. To understand regenerative heterogeneity, we conducted patch-based single-cell RNA sequencing on rare regenerating CST neurons at high depth following PTEN and SOCS3 deletion. Supervised classification with Garnett gave rise to a Regeneration Classifier, which can be broadly applied to predict the regenerative potential of diverse neuronal types across developmental stages or after injury. Network analyses highlighted the importance of antioxidant response and mitochondrial biogenesis. Conditional gene deletion validated a role for NFE2L2 (or NRF2), a master regulator of antioxidant response, in CST regeneration. Our data demonstrate a universal transcriptomic signature underlying the regenerative potential of vastly different neuronal populations and illustrate that deep sequencing of only hundreds of phenotypically identified neurons has the power to advance regenerative biology.


Asunto(s)
Axones , Traumatismos de la Médula Espinal , Humanos , Axones/fisiología , Regeneración Nerviosa/genética , Antioxidantes , Neuronas , Traumatismos de la Médula Espinal/genética , Tractos Piramidales/fisiología , Análisis de la Célula Individual
14.
Cell Death Discov ; 8(1): 390, 2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36123349

RESUMEN

Although protein synthesis is hypothesized to have a pivotal role in axonal repair after central nervous system (CNS) injury, the role of core components of the protein synthesis machinery has not been examined. Notably, some elongation factors possess non-canonical functions that may further impact axonal repair. Here, we examined whether overexpressing eukaryotic elongation factor 1 alpha (eEF1A) proteins enhances the collateral sprouting of corticospinal tract (CST) neurons after unilateral pyramidotomy, along with the underlying molecular mechanisms. We found that overexpressing eEF1A proteins in CST neurons increased the levels of pS6, an indicator for mTOR activity, but not pSTAT3 and pAKT levels, in neuronal somas. Strikingly, overexpressing eEF1A2 alone, but neither eEF1A1 alone nor both factors simultaneously, increased protein synthesis and actin rearrangement in CST neurons. While eEF1A1 overexpression only slightly enhanced CST sprouting after pyramidotomy, eEF1A2 overexpression substantially enhanced this sprouting. Surprisingly, co-overexpression of both eEF1A1 and eEF1A2 led to a sprouting phenotype similar to wild-type controls, suggesting an antagonistic effect of overexpressing both proteins. These data provide the first evidence that overexpressing a core component of the translation machinery, eEF1A2, enhances CST sprouting, likely by a combination of increased protein synthesis, mTOR signaling and actin cytoskeleton rearrangement.

15.
iScience ; 25(11): 105383, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36339257

RESUMEN

Axonal repair is critical for functional recovery after injury of the CNS. We previously reported that neuronal PTEN deletion exhibits an age-dependent decline in promoting axon regeneration from the corticospinal tract (CST). How sprouting of uninjured axons, a naturally occurring form of axonal repair, is impacted by age is unknown. We assessed CST sprouting after unilateral pyramidotomy in PTEN and/or SOCS3-deleted mice at different ages. While PTEN deletion enhances sprouting independently of age, SOCS3 deletion loses its sprouting-promoting effect with age. The synergistic effect of PTEN/SOCS3 co-deletion on CST sprouting is rapidly lost with increased age. Overall, promoting sprouting appears more robust across age than regeneration, yet distinct molecular pathways are differentially impacted by age. Importantly, six-week delayed PTEN deletion promotes CST sprouting across age groups, supporting a clinically relevant time frame for this neural repair strategy independently of age.

16.
J Neurosci ; 30(32): 10899-904, 2010 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-20702718

RESUMEN

After CNS injuries, axon growth inhibitors from the myelin and the scar tissue at the injury site are considered major impediments to axon regeneration. The presence of several classes of inhibitors with multiple members in each class suggests functional redundancy in growth inhibition. To test redundancy within the myelin inhibitory pathway, we analyzed raphe spinal serotonergic (5-HT) axon regeneration in mice deficient in two major myelin inhibitors, Nogo and MAG, and their common receptor NgR1 (or NgR). After a complete transection spinal cord injury, there was no significant enhancement of 5-HT axon regeneration beyond the injury site in either Nogo/MAG/NgR1 triple mutants or NgR1 single mutants. Occasional, genotype-independent traversal of 5-HT axons through GFAP-positive tissue bridges at the injury site implicates GFAP-negative lesion areas as especially inhibitory to 5-HT axons. To assess the contribution of class 3 Semaphorins that are expressed by GFAP-negative meningeal fibroblasts at the injury site, we analyzed mice deficient in PlexinA3 and PlexinA4, two key receptors for class 3 Semaphorins, with or without additional NgR1 deletion. No enhanced regeneration of 5-HT or corticospinal axons was detected in PlexinA3/PlexinA4 double mutants or PlexinA3/PlexinA4/NgR1 triple mutants through a complete transection injury. In contrast with previous reports, these data demonstrate that attenuating myelin or Semaphorin-mediated inhibition of axon growth is insufficient to promote 5-HT axon regeneration and further indicate that even attenuating both classes of inhibitory influences is insufficient to promote regeneration of injured axons through a complete transection spinal cord injury.


Asunto(s)
Axones/fisiología , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Semaforinas/metabolismo , Serotonina/metabolismo , Animales , Axones/patología , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina/deficiencia , Vaina de Mielina/genética , Glicoproteína Asociada a Mielina , Regeneración Nerviosa/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neurregulina-1/deficiencia , Proteínas Nogo , Receptores de Superficie Celular/deficiencia , Semaforinas/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología
17.
Cereb Cortex ; 20(8): 1769-79, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19892785

RESUMEN

The oligodendrocyte myelin glycoprotein is a glycosylphosphatidylinositol-anchored protein expressed by neurons and oligodendrocytes in the central nervous system. Attempts have been made to identify the functions of the myelin-associated inhibitory proteins (MAIPs) after axonal lesion or in neurodegeneration. However, the developmental roles of some of these proteins and their receptors remain elusive. Recent studies indicate that NgR1 and the recently discovered receptor PirB restrict cortical synaptic plasticity. However, the putative factors that trigger these effects are unknown. Because Nogo-A is mostly associated with the endoplasmic reticulum and myelin associated glycoprotein appears late during development, the putative participation of OMgp should be considered. Here, we examine the pattern of development of OMgp immunoreactive elements during mouse telencephalic development. OMgp immunoreactivity in the developing cortex follows the establishment of the thalamo-cortical barrel field. At the cellular level, we located OMgp neuronal membranes in dendrites and axons as well as in brain synaptosome fractions and axon varicosities. Lastly, the analysis of the barrel field in OMgp-deficient mice revealed that although thalamo-cortical connections were formed, their targeting in layer IV was altered, and numerous axons ectopically invaded layers II-III. Our data support the idea that early expressed MAIPs play an active role during development and point to OMgp participating in thalamo-cortical connections.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Glicoproteína Asociada a Mielina/biosíntesis , Glicoproteína Asociada a Mielina/genética , Telencéfalo/metabolismo , Animales , Mapeo Encefálico , Diferenciación Celular/genética , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Proteínas Ligadas a GPI , Conos de Crecimiento/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/deficiencia , Glicoproteína Mielina-Oligodendrócito , Fibras Nerviosas Mielínicas/metabolismo , Vías Nerviosas/embriología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/metabolismo , Ratas , Ratas Sprague-Dawley , Corteza Somatosensorial/citología , Corteza Somatosensorial/embriología , Corteza Somatosensorial/metabolismo , Telencéfalo/citología , Telencéfalo/embriología , Tálamo/citología , Tálamo/embriología , Tálamo/metabolismo
18.
Mol Ther ; 18(8): 1496-500, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20502445

RESUMEN

Adeno-associated virus (AAV) is capable of mediating retrograde viral transduction of central and peripheral neurons. This occurs at a relatively low efficiency, which we previously found to be dependent upon capsid serotype. We sought to augment retrograde transduction by providing increased axonal access to peripherally delivered AAV. Others have described utilizing full transection of peripheral nerves to mediate retrograde viral transduction of motor neurons. Here, we examined the ability of a transient demyelinating event to modulate levels of retrograde AAV transduction. Transient demyelination does not cause lasting functional deficits. Ethidium bromide (EtBr)-induced transient demyelination of the sciatic nerve resulted in significant elevation of retrograde transduction of both motor and sensory neurons. Retrograde transduction levels of motor neurons and heavily myelinated, large-diameter sensory neurons increased at least sixfold following peripheral delivery of self-complementary AAV serotype 1 (scAAV1) and serotype 2 (scAAV2), when preceded by demyelination. These findings identify a means of significantly enhancing retrograde vector transport for use in experimental paradigms requiring either retrograde neuronal identification and gene expression, or translational treatment paradigms.


Asunto(s)
Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo , Dependovirus/genética , Nervio Ciático/efectos de los fármacos , Transducción Genética/métodos , Animales , Etidio/toxicidad , Femenino , Vectores Genéticos/genética , Inmunohistoquímica , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Ratas , Nervio Ciático/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo
19.
Elife ; 102021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33475086

RESUMEN

The conserved MAP3K Dual-Leucine-Zipper Kinase (DLK) and Leucine-Zipper-bearing Kinase (LZK) can activate JNK via MKK4 or MKK7. These two MAP3Ks share similar biochemical activities and undergo auto-activation upon increased expression. Depending on cell-type and nature of insults DLK and LZK can induce pro-regenerative, pro-apoptotic or pro-degenerative responses, although the mechanistic basis of their action is not well understood. Here, we investigated these two MAP3Ks in cerebellar Purkinje cells using loss- and gain-of function mouse models. While loss of each or both kinases does not cause discernible defects in Purkinje cells, activating DLK causes rapid death and activating LZK leads to slow degeneration. Each kinase induces JNK activation and caspase-mediated apoptosis independent of each other. Significantly, deleting CELF2, which regulates alternative splicing of Map2k7, strongly attenuates Purkinje cell degeneration induced by LZK, but not DLK. Thus, controlling the activity levels of DLK and LZK is critical for neuronal survival and health.


Asunto(s)
Quinasas Quinasa Quinasa PAM/genética , Células de Purkinje/metabolismo , Transducción de Señal , Animales , Supervivencia Celular , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones
20.
Genesis ; 48(2): 101-5, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20014422

RESUMEN

Ephrins and Eph receptor tyrosine kinases are cell-surface molecules that serve a multitude of functions in cell-cell communication in development, physiology, and disease. EphA4 is a promiscuous member of the EphA subclass of Eph receptors and can bind to both EphrinAs and EphrinBs. In addition to its well-established roles in guiding the development of neuronal connectivity, EphA4 has been implicated for a role in synaptic plasticity, vascular formation, axon regeneration, and central nervous system repair following injury. However, the study of its role in the adult stage has been hampered by confounding developmental defects in EphA4 germline mutants. Here, we report the generation and molecular characterization of an EphA4 conditional allele along with a novel null allele with a knockin fluorescent reporter gene (mCFP). The conditional allele will be useful in ascertaining postdevelopmental and/or cell type-specific function of EphA4 in physiology, injury, and disease.


Asunto(s)
Alelos , Receptor EphA4/genética , Animales , Comunicación Celular/genética , Femenino , Colorantes Fluorescentes/metabolismo , Marcación de Gen , Genes Reporteros , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Inmunohistoquímica , Indoles/metabolismo , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Genéticos , Mutación , Plasticidad Neuronal , Receptor EphA4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA