Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.266
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(3): 466-468, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36736299

RESUMEN

Microbiota-induced IL-17 production mediates CNS processes and animal behavior. However, its role on the peripheral nervous system (PNS) remains largely unknown. Enamorado et al. demonstrate that commensal-specific Th17 cells are recalled following tissue injury to support local nerve regeneration, a process orchestrated by IL-17 signaling on peripheral neurons.


Asunto(s)
Sistema Nervioso Central , Interleucina-17 , Animales , Sistema Nervioso Periférico , Regeneración Nerviosa/fisiología , Transducción de Señal , Nervios Periféricos , Axones/fisiología
2.
Cell ; 186(3): 607-620.e17, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36640762

RESUMEN

Tissue immunity and responses to injury depend on the coordinated action and communication among physiological systems. Here, we show that, upon injury, adaptive responses to the microbiota directly promote sensory neuron regeneration. At homeostasis, tissue-resident commensal-specific T cells colocalize with sensory nerve fibers within the dermis, express a transcriptional program associated with neuronal interaction and repair, and promote axon growth and local nerve regeneration following injury. Mechanistically, our data reveal that the cytokine interleukin-17A (IL-17A) released by commensal-specific Th17 cells upon injury directly signals to sensory neurons via IL-17 receptor A, the transcription of which is specifically upregulated in injured neurons. Collectively, our work reveals that in the context of tissue damage, preemptive immunity to the microbiota can rapidly bridge biological systems by directly promoting neuronal repair, while also identifying IL-17A as a major determinant of this fundamental process.


Asunto(s)
Interleucina-17 , Microbiota , Regeneración Nerviosa , Células Th17 , Axones , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales , Animales , Ratones , Células Th17/citología
3.
Cell ; 185(1): 77-94, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34995518

RESUMEN

Neurons of the mammalian central nervous system fail to regenerate. Substantial progress has been made toward identifying the cellular and molecular mechanisms that underlie regenerative failure and how altering those pathways can promote cell survival and/or axon regeneration. Here, we summarize those findings while comparing the regenerative process in the central versus the peripheral nervous system. We also highlight studies that advance our understanding of the mechanisms underlying neural degeneration in response to injury, as many of these mechanisms represent primary targets for restoring functional neural circuits.


Asunto(s)
Axones/metabolismo , Sistema Nervioso Central/metabolismo , Regeneración Nerviosa/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Sistema Nervioso Periférico/metabolismo
4.
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
5.
Cell ; 174(3): 521-535.e13, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30033363

RESUMEN

Many human spinal cord injuries are anatomically incomplete but exhibit complete paralysis. It is unknown why spared axons fail to mediate functional recovery in these cases. To investigate this, we undertook a small-molecule screen in mice with staggered bilateral hemisections in which the lumbar spinal cord is deprived of all direct brain-derived innervation, but dormant relay circuits remain. We discovered that a KCC2 agonist restored stepping ability, which could be mimicked by selective expression of KCC2, or hyperpolarizing DREADDs, in the inhibitory interneurons between and around the staggered spinal lesions. Mechanistically, these treatments transformed this injury-induced dysfunctional spinal circuit to a functional state, facilitating the relay of brain-derived commands toward the lumbar spinal cord. Thus, our results identify spinal inhibitory interneurons as a roadblock limiting the integration of descending inputs into relay circuits after injury and suggest KCC2 agonists as promising treatments for promoting functional recovery after spinal cord injury.


Asunto(s)
Traumatismos de la Médula Espinal/tratamiento farmacológico , Simportadores/agonistas , Simportadores/metabolismo , Animales , Axones , Regulación de la Expresión Génica/genética , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración Nerviosa/fisiología , Plasticidad Neuronal/genética , Neuronas/metabolismo , Recuperación de la Función/genética , Recuperación de la Función/fisiología , Médula Espinal , Simportadores/uso terapéutico , Cotransportadores de K Cl
6.
Annu Rev Cell Dev Biol ; 32: 127-141, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27298094

RESUMEN

The brain constantly changes to store memories and adapt to new conditions. One type of plasticity that has gained increasing interest during the last years is the generation of new cells. The generation of both new neurons and glial cells contributes to neural plasticity and to some neural repair. There are substantial differences between mammalian species with regard to the extent of and mechanisms behind cell exchange in neural plasticity. Both neurogenesis and gliogenesis have several specific features in humans, which may contribute to the unique plasticity of the human brain.


Asunto(s)
Regeneración Nerviosa/fisiología , Neurogénesis , Neuroglía/citología , Plasticidad Neuronal/fisiología , Animales , Encéfalo/citología , Humanos , Oligodendroglía/citología
7.
Nat Rev Neurosci ; 25(10): 649-667, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39164450

RESUMEN

Mammalian neurons lose the ability to regenerate their central nervous system axons as they mature during embryonic or early postnatal development. Neuronal maturation requires a transformation from a situation in which neuronal components grow and assemble to one in which these components are fixed and involved in the machinery for effective information transmission and computation. To regenerate after injury, neurons need to overcome this fixed state to reactivate their growth programme. A variety of intracellular processes involved in initiating or sustaining neuronal maturation, including the regulation of gene expression, cytoskeletal restructuring and shifts in intracellular trafficking, have been shown to prevent axon regeneration. Understanding these processes will contribute to the identification of targets to promote repair after injury or disease.


Asunto(s)
Axones , Regeneración Nerviosa , Neurogénesis , Animales , Regeneración Nerviosa/fisiología , Axones/fisiología , Humanos , Neurogénesis/fisiología , Neuronas/fisiología
8.
Annu Rev Neurosci ; 42: 209-226, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-30883262

RESUMEN

How the nervous system is wired has been a central question of neuroscience since the inception of the field, and many of the foundational discoveries and conceptual advances have been made through the study of invertebrate experimental organisms, including Caenorhabditis elegans and Drosophila melanogaster. Although many guidance molecules and receptors have been identified, recent experiments have shed light on the many modes of action for these pathways. Here, we summarize the recent progress in determining how the physical and temporal constraints of the surrounding environment provide instructive regulations in nervous system wiring. We use Netrin and its receptors as an example to analyze the complexity of how they guide neurite outgrowth. In neurite repair, conserved injury detection and response-signaling pathways regulate gene expression and cytoskeletal dynamics. We also describe recent developments in the research on molecular mechanisms of neurite regeneration in worms and flies.


Asunto(s)
Caenorhabditis elegans/fisiología , Drosophila melanogaster/fisiología , Regeneración Nerviosa/fisiología , Neurogénesis , Proyección Neuronal/fisiología , Animales , Orientación del Axón/fisiología , Caenorhabditis elegans/citología , Caenorhabditis elegans/crecimiento & desarrollo , Señalización del Calcio , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Larva , Sistema de Señalización de MAP Quinasas/fisiología , Microtúbulos/fisiología , Receptores de Netrina/fisiología , Netrinas/fisiología , Fosfatidilserinas/fisiología , Factores de Tiempo , Traumatismos del Sistema Nervioso/fisiopatología
9.
Development ; 151(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39007397

RESUMEN

Many genes are known to regulate retinal regeneration after widespread tissue damage. Conversely, genes controlling regeneration after limited cell loss, as per degenerative diseases, are undefined. As stem/progenitor cell responses scale to injury levels, understanding how the extent and specificity of cell loss impact regenerative processes is important. Here, transgenic zebrafish enabling selective retinal ganglion cell (RGC) ablation were used to identify genes that regulate RGC regeneration. A single cell multiomics-informed screen of 100 genes identified seven knockouts that inhibited and 11 that promoted RGC regeneration. Surprisingly, 35 out of 36 genes known and/or implicated as being required for regeneration after widespread retinal damage were not required for RGC regeneration. The loss of seven even enhanced regeneration kinetics, including the proneural factors neurog1, olig2 and ascl1a. Mechanistic analyses revealed that ascl1a disruption increased the propensity of progenitor cells to produce RGCs, i.e. increased 'fate bias'. These data demonstrate plasticity in the mechanism through which Müller glia convert to a stem-like state and context specificity in how genes function during regeneration. Increased understanding of how the regeneration of disease-relevant cell types is specifically controlled will support the development of disease-tailored regenerative therapeutics.


Asunto(s)
Animales Modificados Genéticamente , Células Ganglionares de la Retina , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sistemas CRISPR-Cas/genética , Regeneración/genética , Regeneración/fisiología , Retina/metabolismo , Retina/citología , Células Madre/metabolismo , Células Madre/citología , Factores de Transcripción
10.
Proc Natl Acad Sci U S A ; 121(28): e2400596121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38968119

RESUMEN

In adult songbirds, new neurons are born in large numbers in the proliferative ventricular zone in the telencephalon and migrate to the adjacent song control region HVC (acronym used as proper name) [A. Reiner et al., J. Comp. Neurol. 473, 377-414 (2004)]. Many of these new neurons send long axonal projections to the robust nucleus of the arcopallium (RA). The HVC-RA circuit is essential for producing stereotyped learned song. The function of adult neurogenesis in this circuit has not been clear. A previous study suggested that it is important for the production of well-structured songs [R. E. Cohen, M. Macedo-Lima, K. E. Miller, E. A. Brenowitz, J. Neurosci. 36, 8947-8956 (2016)]. We tested this hypothesis by infusing the neuroblast migration inhibitor cyclopamine into HVC of male Gambel's white-crowned sparrows (Zonotrichia leucophrys gambelii) to block seasonal regeneration of the HVC-RA circuit. Decreasing the number of new neurons in HVC prevented both the increase in spontaneous electrical activity of RA neurons and the improved structure of songs that would normally occur as sparrows enter breeding condition. These results show that the incorporation of new neurons into the adult HVC is necessary for the recovery of both electrical activity and song behavior in breeding birds and demonstrate the value of the bird song system as a model for investigating adult neurogenesis at the level of long projection neural circuits.


Asunto(s)
Neurogénesis , Prosencéfalo , Vocalización Animal , Animales , Neurogénesis/fisiología , Prosencéfalo/fisiología , Prosencéfalo/citología , Vocalización Animal/fisiología , Masculino , Gorriones/fisiología , Neuronas/fisiología , Regeneración Nerviosa/fisiología
11.
PLoS Genet ; 20(7): e1011367, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39058749

RESUMEN

The pathway for axon regeneration in Caenorhabditis elegans is activated by SVH-1, a growth factor belonging to the HGF/plasminogen family. SVH-1 is a dual-function factor that acts as an HGF-like growth factor to promote axon regeneration and as a protease to regulate early development. It is important to understand how SVH-1 is converted from a protease to a growth factor for axon regeneration. In this study, we demonstrate that cytidine deaminase (CDD) SVH-17/CDD-2 plays a role in the functional conversion of SVH-1. We find that the codon exchange of His-755 to Tyr in the Asp-His-Ser catalytic triad of SVH-1 can suppress the cdd-2 defect in axon regeneration. Furthermore, the stem hairpin structure around the His-755 site in svh-1 mRNA is required for the activation of axon regeneration by SVH-1. These results suggest that CDD-2 promotes axon regeneration by transforming the function of SVH-1 from a protease to a growth factor through modification of svh-1 mRNA.


Asunto(s)
Axones , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Citidina Desaminasa , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Axones/metabolismo , Axones/fisiología , Citidina Desaminasa/metabolismo , Citidina Desaminasa/genética , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/genética , Regeneración/genética
12.
Proc Natl Acad Sci U S A ; 121(6): e2305947121, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38289952

RESUMEN

Optic neuropathies, characterized by injury of retinal ganglion cell (RGC) axons of the optic nerve, cause incurable blindness worldwide. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) represent a promising "cell-free" therapy for regenerative medicine; however, the therapeutic effect on neural restoration fluctuates, and the underlying mechanism is poorly understood. Here, we illustrated that intraocular administration of MSC-sEVs promoted both RGC survival and axon regeneration in an optic nerve crush mouse model. Mechanistically, MSC-sEVs primarily targeted retinal mural cells to release high levels of colony-stimulating factor 3 (G-CSF) that recruited a neural restorative population of Ly6Clow monocytes/monocyte-derived macrophages (Mo/MΦ). Intravitreal administration of G-CSF, a clinically proven agent for treating neutropenia, or donor Ly6Clow Mo/MΦ markedly improved neurological outcomes in vivo. Together, our data define a unique mechanism of MSC-sEV-induced G-CSF-to-Ly6Clow Mo/MΦ signaling in repairing optic nerve injury and highlight local delivery of MSC-sEVs, G-CSF, and Ly6Clow Mo/MΦ as therapeutic paradigms for the treatment of optic neuropathies.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Traumatismos del Nervio Óptico , Ratones , Animales , Axones/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/terapia , Traumatismos del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/fisiología , Células Madre Mesenquimatosas/metabolismo , Vesículas Extracelulares/metabolismo , Macrófagos/metabolismo
13.
Proc Natl Acad Sci U S A ; 121(31): e2315599121, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39058581

RESUMEN

Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing "synaptopathy" and hearing loss. Cocultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that, when P3-5 denervated organs of Corti are cocultured with SGNs, newly formed IHC/SGN synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When using older organs of Corti at P10-11, synaptic activity probed by deconvolution showed more mature release properties, closer to the specialized mode of IHC synaptic transmission crucial for coding the sound signal. This functional assessment of newly formed IHC synapses developed here, provides a powerful tool for testing approaches to improve synapse regeneration.


Asunto(s)
Ganglio Espiral de la Cóclea , Sinapsis , Animales , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/fisiología , Sinapsis/fisiología , Ratones , Células Ciliadas Auditivas Internas/fisiología , Células Ciliadas Auditivas Internas/metabolismo , Transmisión Sináptica/fisiología , Neuronas/fisiología , Neuronas/metabolismo , Regeneración/fisiología , Células Ciliadas Auditivas/fisiología , Técnicas de Cocultivo/métodos , Optogenética/métodos , Regeneración Nerviosa/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Órgano Espiral/fisiología , Órgano Espiral/citología , Órgano Espiral/metabolismo
14.
Proc Natl Acad Sci U S A ; 121(38): e2402518121, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39254997

RESUMEN

The in vivo three-dimensional genomic architecture of adult mature neurons at homeostasis and after medically relevant perturbations such as axonal injury remains elusive. Here, we address this knowledge gap by mapping the three-dimensional chromatin architecture and gene expression program at homeostasis and after sciatic nerve injury in wild-type and cohesin-deficient mouse sensory dorsal root ganglia neurons via combinatorial Hi-C, promoter-capture Hi-C, CUT&Tag for H3K27ac and RNA-seq. We find that genes involved in axonal regeneration form long-range, complex chromatin loops, and that cohesin is required for the full induction of the regenerative transcriptional program. Importantly, loss of cohesin results in disruption of chromatin architecture and severely impaired nerve regeneration. Complex enhancer-promoter loops are also enriched in the human fetal cortical plate, where the axonal growth potential is highest, and are lost in mature adult neurons. Together, these data provide an original three-dimensional chromatin map of adult sensory neurons in vivo and demonstrate a role for cohesin-dependent long-range promoter interactions in nerve regeneration.


Asunto(s)
Axones , Cromatina , Cohesinas , Regeneración Nerviosa , Regiones Promotoras Genéticas , Células Receptoras Sensoriales , Animales , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Ratones , Regiones Promotoras Genéticas/genética , Cromatina/metabolismo , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Axones/metabolismo , Axones/fisiología , Humanos , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Elementos de Facilitación Genéticos/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ganglios Espinales/metabolismo , Ganglios Espinales/citología , Nervio Ciático/metabolismo
15.
Proc Natl Acad Sci U S A ; 121(39): e2404395121, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39292743

RESUMEN

Adult central nervous system (CNS) neurons down-regulate growth programs after injury, leading to persistent regeneration failure. Coordinated lipids metabolism is required to synthesize membrane components during axon regeneration. However, lipids also function as cell signaling molecules. Whether lipid signaling contributes to axon regeneration remains unclear. In this study, we showed that lipin1 orchestrates mechanistic target of rapamycin (mTOR) and STAT3 signaling pathways to determine axon regeneration. We established an mTOR-lipin1-phosphatidic acid/lysophosphatidic acid-mTOR loop that acts as a positive feedback inhibitory signaling, contributing to the persistent suppression of CNS axon regeneration following injury. In addition, lipin1 knockdown (KD) enhances corticospinal tract (CST) sprouting after unilateral pyramidotomy and promotes CST regeneration following complete spinal cord injury (SCI). Furthermore, lipin1 KD enhances sensory axon regeneration after SCI. Overall, our research reveals that lipin1 functions as a central regulator to coordinate mTOR and STAT3 signaling pathways in the CNS neurons and highlights the potential of lipin1 as a promising therapeutic target for promoting the regeneration of motor and sensory axons after SCI.


Asunto(s)
Axones , Neuronas Motoras , Regeneración Nerviosa , Fosfatidato Fosfatasa , Factor de Transcripción STAT3 , Transducción de Señal , Traumatismos de la Médula Espinal , Serina-Treonina Quinasas TOR , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/genética , Animales , Axones/metabolismo , Axones/fisiología , Regeneración Nerviosa/fisiología , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Fosfatidato Fosfatasa/metabolismo , Fosfatidato Fosfatasa/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/fisiología , Ratones , Ácidos Fosfatidicos/metabolismo , Células Receptoras Sensoriales/metabolismo , Femenino , Tractos Piramidales/metabolismo , Tractos Piramidales/patología
16.
PLoS Genet ; 20(8): e1011388, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39186815

RESUMEN

Most neurons are not replaced after injury and thus possess robust intrinsic mechanisms for repair after damage. Axon injury triggers a calcium wave, and calcium and cAMP can augment axon regeneration. In comparison to axon regeneration, dendrite regeneration is poorly understood. To test whether calcium and cAMP might also be involved in dendrite injury signaling, we tracked the responses of Drosophila dendritic arborization neurons to laser severing of axons and dendrites. We found that calcium and subsequently cAMP accumulate in the cell body after both dendrite and axon injury. Two voltage-gated calcium channels (VGCCs), L-Type and T-Type, are required for the calcium influx in response to dendrite injury and play a role in rapid initiation of dendrite regeneration. The AC8 family adenylyl cyclase, Ac78C, is required for cAMP production after dendrite injury and timely initiation of regeneration. Injury-induced cAMP production is sensitive to VGCC reduction, placing calcium upstream of cAMP generation. We propose that two VGCCs initiate global calcium influx in response to dendrite injury followed by production of cAMP by Ac78C. This signaling pathway promotes timely initiation of dendrite regrowth several hours after dendrite damage.


Asunto(s)
Adenilil Ciclasas , Canales de Calcio Tipo L , Calcio , AMP Cíclico , Dendritas , Animales , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/genética , Axones/metabolismo , Axones/fisiología , Calcio/metabolismo , Canales de Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo T/metabolismo , Canales de Calcio Tipo T/genética , Señalización del Calcio/genética , AMP Cíclico/metabolismo , Dendritas/metabolismo , Drosophila/genética , Drosophila melanogaster/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Regeneración Nerviosa/fisiología , Regeneración Nerviosa/genética , Neuronas/metabolismo , Regeneración/genética , Regeneración/fisiología , Transducción de Señal
17.
EMBO J ; 41(20): e110486, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36004759

RESUMEN

The proteasome is essential for cellular responses to various physiological stressors. However, how proteasome function impacts the stress resilience of regenerative damaged motor neurons remains unclear. Here, we develop a unique mouse model using a regulatory element of the activating transcription factor (Atf3) gene to label mitochondria in a damage-induced manner while simultaneously genetically disrupting the proteasome. Using this model, we observed that in injury-induced proteasome-deficient mouse motor neurons, the increase of mitochondrial influx from soma into axons is inhibited because neurons fail to disassemble ankyrin G, an organizer of the axon initial segment (AIS), in a proteasome-dependent manner. Further, these motor neurons exhibit amyotrophic lateral sclerosis (ALS)-like degeneration despite having regenerative potential. Selectively vulnerable motor neurons in SOD1G93A ALS mice, which induce ATF3 in response to pathological damage, also fail to disrupt the AIS, limiting the number of axonal mitochondria at a pre-symptomatic stage. Thus, damage-induced proteasome-sensitive AIS disassembly could be a critical post-translational response for damaged motor neurons to temporarily transit to an immature state and meet energy demands for axon regeneration or preservation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Segmento Inicial del Axón , Esclerosis Amiotrófica Lateral/patología , Animales , Ancirinas/metabolismo , Axones/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/patología , Neuronas Motoras/metabolismo , Regeneración Nerviosa/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Superóxido Dismutasa-1/genética
18.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37102704

RESUMEN

The neurons of our central nervous system (CNS) are unable to regenerate axons following injury, which can result in permanent damage. A new paper in Development demonstrates that newly formed oligodendrocytes contribute to axon regeneration inhibition. To hear more about the story, we caught up with first authors, Jian Xing, Agnieszka Lukomska and Bruce Rheaume, and the corresponding author Ephraim Trakhtenberg, Assistant Professor at the University of Connecticut (UConn) School of Medicine.


Asunto(s)
Axones , Regeneración Nerviosa , Axones/fisiología , Regeneración Nerviosa/fisiología , Neuronas , Sistema Nervioso Central , Oligodendroglía
19.
Development ; 150(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37997694

RESUMEN

Identification of signaling events that contribute to innate spinal cord regeneration in zebrafish can uncover new targets for modulating injury responses of the mammalian central nervous system. Using a chemical screen, we identify JNK signaling as a necessary regulator of glial cell cycling and tissue bridging during spinal cord regeneration in larval zebrafish. With a kinase translocation reporter, we visualize and quantify JNK signaling dynamics at single-cell resolution in glial cell populations in developing larvae and during injury-induced regeneration. Glial JNK signaling is patterned in time and space during development and regeneration, decreasing globally as the tissue matures and increasing in the rostral cord stump upon transection injury. Thus, dynamic and regional regulation of JNK signaling help to direct glial cell behaviors during innate spinal cord regeneration.


Asunto(s)
Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Animales , Larva , Mamíferos , Regeneración Nerviosa/fisiología , Neuroglía/fisiología , Médula Espinal , Pez Cebra/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos
20.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36971369

RESUMEN

Failure of central nervous system projection neurons to spontaneously regenerate long-distance axons underlies irreversibility of white matter pathologies. A barrier to axonal regenerative research is that the axons regenerating in response to experimental treatments stall growth before reaching post-synaptic targets. Here, we test the hypothesis that the interaction of regenerating axons with live oligodendrocytes, which were absent during developmental axon growth, contributes to stalling axonal growth. To test this hypothesis, first, we used single cell RNA-seq (scRNA-seq) and immunohistology to investigate whether post-injury born oligodendrocytes incorporate into the glial scar after optic nerve injury. Then, we administered demyelination-inducing cuprizone and stimulated axon regeneration by Pten knockdown (KD) after optic nerve crush. We found that post-injury born oligodendrocyte lineage cells incorporate into the glial scar, where they are susceptible to the demyelination diet, which reduced their presence in the glial scar. We further found that the demyelination diet enhanced Pten KD-stimulated axon regeneration and that localized cuprizone injection promoted axon regeneration. We also present a resource for comparing the gene expression of scRNA-seq-profiled normal and injured optic nerve oligodendrocyte lineage cells.


Asunto(s)
Axones , Enfermedades Desmielinizantes , Humanos , Axones/fisiología , Gliosis/metabolismo , Gliosis/patología , Cuprizona , Regeneración Nerviosa/fisiología , Células Ganglionares de la Retina/metabolismo , Oligodendroglía , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA