Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 155(4): 894-908, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24209626

RESUMEN

Reactivation of a silent transcriptional program is a critical step in successful axon regeneration following injury. Yet how such a program is unlocked after injury remains largely unexplored. We found that axon injury in peripheral sensory neurons elicits a back-propagating calcium wave that invades the soma and causes nuclear export of HDAC5 in a PKCµ-dependent manner. Injury-induced HDAC5 nuclear export enhances histone acetylation to activate a proregenerative gene-expression program. HDAC5 nuclear export is required for axon regeneration, as expression of a nuclear-trapped HDAC5 mutant prevents axon regeneration, whereas enhancing HDAC5 nuclear export promotes axon regeneration in vitro and in vivo. Components of this HDAC5 pathway failed to be activated in a model of central nervous system injury. These studies reveal a signaling mechanism from the axon injury site to the soma that controls neuronal growth competence and suggest a role for HDAC5 as a transcriptional switch controlling axon regeneration.


Asunto(s)
Transporte Activo de Núcleo Celular , Axones/fisiología , Histona Desacetilasas/metabolismo , Células Receptoras Sensoriales/fisiología , Transcripción Genética , Animales , Señalización del Calcio , Histona Desacetilasas/genética , Ratones , Mutación , Regeneración Nerviosa , Transducción de Señal
2.
Proc Natl Acad Sci U S A ; 120(7): e2215906120, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36763532

RESUMEN

Sensory neurons located in dorsal root ganglia (DRG) convey sensory information from peripheral tissue to the brain. After peripheral nerve injury, sensory neurons switch to a regenerative state to enable axon regeneration and functional recovery. This process is not cell autonomous and requires glial and immune cells. Macrophages in the DRG (DRGMacs) accumulate in response to nerve injury, but their origin and function remain unclear. Here, we mapped the fate and response of DRGMacs to nerve injury using macrophage depletion, fate-mapping, and single-cell transcriptomics. We identified three subtypes of DRGMacs after nerve injury in addition to a small population of circulating bone-marrow-derived precursors. Self-renewing macrophages, which proliferate from local resident macrophages, represent the largest population of DRGMacs. The other two subtypes include microglia-like cells and macrophage-like satellite glial cells (SGCs) (Imoonglia). We show that self-renewing DRGMacs contribute to promote axon regeneration. Using single-cell transcriptomics data and CellChat to simulate intercellular communication, we reveal that macrophages express the neuroprotective and glioprotective ligand prosaposin and communicate with SGCs via the prosaposin receptor GPR37L1. These data highlight that DRGMacs have the capacity to self-renew, similarly to microglia in the Central nervous system (CNS) and contribute to promote axon regeneration. These data also reveal the heterogeneity of DRGMacs and their potential neuro- and glioprotective roles, which may inform future therapeutic approaches to treat nerve injury.


Asunto(s)
Axones , Traumatismos de los Nervios Periféricos , Humanos , Axones/fisiología , Regeneración Nerviosa/fisiología , Ganglios Espinales/fisiología , Macrófagos/fisiología , Neuroglía , Receptores Acoplados a Proteínas G/genética
4.
J Biol Chem ; 298(3): 101647, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35101451

RESUMEN

The dual leucine zipper kinase (DLK) is a key regulator of axon regeneration and degeneration in response to neuronal injury; however, regulatory mechanisms of the DLK function via its interacting proteins are largely unknown. To better understand the molecular mechanism of DLK function, we performed yeast two-hybrid screening analysis and identified FK506-binding protein-like (FKBPL, also known as WAF-1/CIP1 stabilizing protein 39) as a DLK-binding protein. FKBPL binds to the kinase domain of DLK and inhibits its kinase activity. In addition, FKBPL induces DLK protein degradation through ubiquitin-dependent pathways. We further assessed other members in the FKBP protein family and found that FK506-binding protein 8 (FKBP8) also induced DLK degradation. We identified the lysine 271 residue in the kinase domain as a major site of DLK ubiquitination and SUMO3 conjugation and was thus responsible for regulating FKBP8-mediated proteasomal degradation that was inhibited by the substitution of the lysine 271 to arginine. FKBP8-mediated degradation of DLK is mediated by autophagy pathway because knockdown of Atg5 inhibited DLK destabilization. We show that in vivo overexpression of FKBP8 delayed the progression of axon degeneration and suppressed neuronal death after axotomy in sciatic and optic nerves. Taken together, this study identified FKBPL and FKBP8 as novel DLK-interacting proteins that regulate DLK stability via the ubiquitin-proteasome and lysosomal protein degradation pathways.


Asunto(s)
Axones , Quinasas Quinasa Quinasa PAM , Degeneración Nerviosa , Proteínas de Unión a Tacrolimus , Axones/enzimología , Axones/metabolismo , Axones/patología , Leucina Zippers , Lisina/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Regeneración Nerviosa , Proteínas de Unión a Tacrolimus/metabolismo , Ubiquitina/metabolismo
5.
Nat Rev Neurosci ; 19(6): 323-337, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29666508

RESUMEN

Permanent disabilities following CNS injuries result from the failure of injured axons to regenerate and rebuild functional connections with their original targets. By contrast, injury to peripheral nerves is followed by robust regeneration, which can lead to recovery of sensory and motor functions. This regenerative response requires the induction of widespread transcriptional and epigenetic changes in injured neurons. Considerable progress has been made in recent years in understanding how peripheral axon injury elicits these widespread changes through the coordinated actions of transcription factors, epigenetic modifiers and, to a lesser extent, microRNAs. Although many questions remain about the interplay between these mechanisms, these new findings provide important insights into the pivotal role of coordinated gene expression and chromatin remodelling in the neuronal response to injury.


Asunto(s)
Axones/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Regeneración Nerviosa , Neuronas/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Expresión Génica , Humanos , MicroARNs/metabolismo , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/metabolismo , Transducción de Señal
6.
J Allergy Clin Immunol ; 149(4): 1473-1480.e6, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34560104

RESUMEN

BACKGROUND: Chronic pruritus, or itch, is common and debilitating, but the neuroimmune mechanisms that drive chronic itch are only starting to be elucidated. Recent studies demonstrate that the IL-33 receptor (IL-33R) is expressed by sensory neurons. However, whether sensory neuron-restricted activity of IL-33 is necessary for chronic itch remains poorly understood. OBJECTIVES: We sought to determine if IL-33 signaling in sensory neurons is critical for the development of chronic itch in 2 divergent pruritic disease models. METHODS: Plasma levels of IL-33 were assessed in patients with atopic dermatitis (AD) and chronic pruritus of unknown origin (CPUO). Mice were generated to conditionally delete IL-33R from sensory neurons. The contribution of neuronal IL-33R signaling to chronic itch development was tested in mouse models that recapitulate key pathologic features of AD and CPUO, respectively. RESULTS: IL-33 was elevated in both AD and CPUO as well as their respective mouse models. While neuron-restricted IL-33R signaling was dispensable for itch in AD-like disease, it was required for the development of dry skin itch in a mouse model that mirrors key aspects of CPUO pathology. CONCLUSIONS: These data highlight how IL-33 may be a predominant mediator of itch in certain contexts, depending on the tissue microenvironment. Further, this study provides insight into future therapeutic strategies targeting the IL-33 pathway for chronic itch.


Asunto(s)
Dermatitis Atópica , Interleucina-33 , Animales , Modelos Animales de Enfermedad , Humanos , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33/metabolismo , Ratones , Prurito , Células Receptoras Sensoriales/metabolismo , Transducción de Señal , Piel
7.
J Lipid Res ; 62: 100079, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33894211

RESUMEN

Vascular disease contributes to neurodegeneration, which is associated with decreased blood pressure in older humans. Plasmalogens, ether phospholipids produced by peroxisomes, are decreased in Alzheimer's disease, Parkinson's disease, and other neurodegenerative disorders. However, the mechanistic links between ether phospholipids, blood pressure, and neurodegeneration are not fully understood. Here, we show that endothelium-derived ether phospholipids affect blood pressure, behavior, and neurodegeneration in mice. In young adult mice, inducible endothelial-specific disruption of PexRAP, a peroxisomal enzyme required for ether lipid synthesis, unexpectedly decreased circulating plasmalogens. PexRAP endothelial knockout (PEKO) mice responded normally to hindlimb ischemia but had lower blood pressure and increased plasma renin activity. In PEKO as compared with control mice, tyrosine hydroxylase was decreased in the locus coeruleus, which maintains blood pressure and arousal. PEKO mice moved less, slept more, and had impaired attention to and recall of environmental events as well as mild spatial memory deficits. In PEKO hippocampus, gliosis was increased, and a plasmalogen associated with memory was decreased. Despite lower blood pressure, PEKO mice had generally normal homotopic functional connectivity by optical neuroimaging of the cerebral cortex. Decreased glycogen synthase kinase-3 phosphorylation, a marker of neurodegeneration, was detected in PEKO cerebral cortex. In a co-culture system, PexRAP knockdown in brain endothelial cells decreased glycogen synthase kinase-3 phosphorylation in co-cultured astrocytes that was rescued by incubation with the ether lipid alkylglycerol. Taken together, our findings suggest that endothelium-derived ether lipids mediate several biological processes and may also confer neuroprotection in mice.


Asunto(s)
Presión Sanguínea
8.
Proc Natl Acad Sci U S A ; 115(52): E12417-E12426, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30530687

RESUMEN

Injured peripheral sensory neurons switch to a regenerative state after axon injury, which requires transcriptional and epigenetic changes. However, the roles and mechanisms of gene inactivation after injury are poorly understood. Here, we show that DNA methylation, which generally leads to gene silencing, is required for robust axon regeneration after peripheral nerve lesion. Ubiquitin-like containing PHD ring finger 1 (UHRF1), a critical epigenetic regulator involved in DNA methylation, increases upon axon injury and is required for robust axon regeneration. The increased level of UHRF1 results from a decrease in miR-9. The level of another target of miR-9, the transcriptional regulator RE1 silencing transcription factor (REST), transiently increases after injury and is required for axon regeneration. Mechanistically, UHRF1 interacts with DNA methyltransferases (DNMTs) and H3K9me3 at the promoter region to repress the expression of the tumor suppressor gene phosphatase and tensin homolog (PTEN) and REST. Our study reveals an epigenetic mechanism that silences tumor suppressor genes and restricts REST expression in time after injury to promote axon regeneration.


Asunto(s)
Regeneración Nerviosa/genética , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Animales , Axones/metabolismo , Axones/fisiología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Metilación de ADN/genética , Epigénesis Genética/genética , Epigenómica/métodos , Femenino , Regulación de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen/fisiología , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración Nerviosa/fisiología , Regiones Promotoras Genéticas/genética , Proteínas Represoras/metabolismo , Nervio Ciático/lesiones , Ubiquitina-Proteína Ligasas
9.
J Neurosci ; 39(1): 28-43, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389838

RESUMEN

Neuronal hyperexcitability is one of the major characteristics of fragile X syndrome (FXS), yet the molecular mechanisms of this critical dysfunction remain poorly understood. Here we report a major role of voltage-independent potassium (K+)-channel dysfunction in hyperexcitability of CA3 pyramidal neurons in Fmr1 knock-out (KO) mice. We observed a reduction of voltage-independent small conductance calcium (Ca2+)-activated K+ (SK) currents in both male and female mice, leading to decreased action potential (AP) threshold and reduced medium afterhyperpolarization. These SK-channel-dependent deficits led to markedly increased AP firing and abnormal input-output signal transmission of CA3 pyramidal neurons. The SK-current defect was mediated, at least in part, by loss of FMRP interaction with the SK channels (specifically the SK2 isoform), without changes in channel expression. Intracellular application of selective SK-channel openers or a genetic reintroduction of an N-terminal FMRP fragment lacking the ability to associate with polyribosomes normalized all observed excitability defects in CA3 pyramidal neurons of Fmr1 KO mice. These results suggest that dysfunction of voltage-independent SK channels is the primary cause of CA3 neuronal hyperexcitability in Fmr1 KO mice and support the critical translation-independent role for the fragile X mental retardation protein as a regulator of neural excitability. Our findings may thus provide a new avenue to ameliorate hippocampal excitability defects in FXS.SIGNIFICANCE STATEMENT Despite two decades of research, no effective treatment is currently available for fragile X syndrome (FXS). Neuronal hyperexcitability is widely considered one of the hallmarks of FXS. Excitability research in the FXS field has thus far focused primarily on voltage-gated ion channels, while contributions from voltage-independent channels have been largely overlooked. Here we report that voltage-independent small conductance calcium-activated potassium (SK)-channel dysfunction causes hippocampal neuron hyperexcitability in the FXS mouse model. Our results support the idea that translation-independent function of fragile X mental retardation protein has a major role in regulating ion-channel activity, specifically the SK channels, in hyperexcitability defects in FXS. Our findings may thus open a new direction to ameliorate hippocampal excitability defects in FXS.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/fisiología , Hipocampo/fisiología , Neuronas/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Potenciales de Acción/fisiología , Animales , Región CA3 Hipocampal/citología , Región CA3 Hipocampal/fisiología , Femenino , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musgosas del Hipocampo/fisiología , Células Piramidales/fisiología , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Transmisión Sináptica/fisiología
10.
J Biol Chem ; 294(44): 16374-16384, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31527079

RESUMEN

Microtubules are cytoskeletal polymers that perform diverse cellular functions. The plus ends of microtubules promote polymer assembly and disassembly and connect the microtubule tips to other cellular structures. The dynamics and functions of microtubule plus ends are governed by microtubule plus end-tracking proteins (+TIPs). Here we report that the Arabidopsis thaliana SPIRAL1 (SPR1) protein, which regulates directional cell expansion, is an autonomous +TIP. Using in vitro reconstitution experiments and total internal reflection fluorescence microscopy, we demonstrate that the conserved N-terminal region of SPR1 and its GGG motif are necessary for +TIP activity whereas the conserved C-terminal region and its PGGG motif are not. We further show that the N- and C-terminal regions, either separated or when fused in tandem (NC), are sufficient for +TIP activity and do not significantly perturb microtubule plus-end dynamics compared with full-length SPR1. We also found that exogenously expressed SPR1-GFP and NC-GFP label microtubule plus ends in plant and animal cells. These results establish SPR1 as a new type of intrinsic +TIP and reveal the utility of NC-GFP as a versatile microtubule plus-end marker.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Microtúbulos/metabolismo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Plantas/metabolismo , Unión Proteica
11.
Mol Cell Proteomics ; 15(2): 382-93, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26297514

RESUMEN

Neurons are extremely polarized cells. Axon lengths often exceed the dimension of the neuronal cell body by several orders of magnitude. These extreme axonal lengths imply that neurons have mastered efficient mechanisms for long distance signaling between soma and synaptic terminal. These elaborate mechanisms are required for neuronal development and maintenance of the nervous system. Neurons can fine-tune long distance signaling through calcium wave propagation and bidirectional transport of proteins, vesicles, and mRNAs along microtubules. The signal transmission over extreme lengths also ensures that information about axon injury is communicated to the soma and allows for repair mechanisms to be engaged. This review focuses on the different mechanisms employed by neurons to signal over long axonal distances and how signals are interpreted in the soma, with an emphasis on proteomic studies. We also discuss how proteomic approaches could help further deciphering the signaling mechanisms operating over long distance in axons.


Asunto(s)
Axones/metabolismo , Sistema Nervioso/metabolismo , Neuronas/metabolismo , Proteómica , Axones/patología , Señalización del Calcio , Polaridad Celular/genética , Humanos , Sistema Nervioso/crecimiento & desarrollo , Neuronas/patología , Transmisión Sináptica/genética
12.
Proc Natl Acad Sci U S A ; 112(4): 949-56, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25561520

RESUMEN

Fragile X syndrome (FXS) results in intellectual disability (ID) most often caused by silencing of the fragile X mental retardation 1 (FMR1) gene. The resulting absence of fragile X mental retardation protein 1 (FMRP) leads to both pre- and postsynaptic defects, yet whether the pre- and postsynaptic functions of FMRP are independent and have distinct roles in FXS neuropathology remain poorly understood. Here, we demonstrate an independent presynaptic function for FMRP through the study of an ID patient with an FMR1 missense mutation. This mutation, c.413G > A (R138Q), preserves FMRP's canonical functions in RNA binding and translational regulation, which are traditionally associated with postsynaptic compartments. However, neuronally driven expression of the mutant FMRP is unable to rescue structural defects at the neuromuscular junction in fragile x mental retardation 1 (dfmr1)-deficient Drosophila, suggesting a presynaptic-specific impairment. Furthermore, mutant FMRP loses the ability to rescue presynaptic action potential (AP) broadening in Fmr1 KO mice. The R138Q mutation also disrupts FMRP's interaction with the large-conductance calcium-activated potassium (BK) channels that modulate AP width. These results reveal a presynaptic- and translation-independent function of FMRP that is linked to a specific subset of FXS phenotypes.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil , Mutación Missense , Convulsiones , Potenciales de Acción/genética , Sustitución de Aminoácidos , Animales , Niño , Preescolar , Drosophila , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Síndrome del Cromosoma X Frágil/fisiopatología , Regulación de la Expresión Génica/genética , Humanos , Masculino , Ratones , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología , Convulsiones/fisiopatología
13.
J Neurosci ; 36(49): 12351-12367, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27927955

RESUMEN

Schwann cells (SCs) are essential for proper peripheral nerve development and repair, although the mechanisms regulating these processes are incompletely understood. We previously showed that the adhesion G protein-coupled receptor Gpr126/Adgrg6 is essential for SC development and myelination. Interestingly, the expression of Gpr126 is maintained in adult SCs, suggestive of a function in the mature nerve. We therefore investigated the role of Gpr126 in nerve repair by studying an inducible SC-specific Gpr126 knock-out mouse model. Here, we show that remyelination is severely delayed after nerve-crush injury. Moreover, we also observe noncell-autonomous defects in macrophage recruitment and axon regeneration in injured nerves following loss of Gpr126 in SCs. This work demonstrates that Gpr126 has critical SC-autonomous and SC-nonautonomous functions in remyelination and peripheral nerve repair. SIGNIFICANCE STATEMENT: Lack of robust remyelination represents one of the major barriers to recovery of neurological functions in disease or following injury in many disorders of the nervous system. Here we show that the adhesion class G protein-coupled receptor (GPCR) Gpr126/Adgrg6 is required for remyelination, macrophage recruitment, and axon regeneration following nerve injury. At least 30% of all approved drugs target GPCRs; thus, Gpr126 represents an attractive potential target to stimulate repair in myelin disease or following nerve injury.


Asunto(s)
Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/patología , Receptores Acoplados a Proteínas G/genética , Células de Schwann/patología , Animales , Axones , Ratones , Ratones Noqueados , Músculo Esquelético/inervación , Músculo Esquelético/patología , Vaina de Mielina , Compresión Nerviosa , Regeneración Nerviosa , Infiltración Neutrófila , Nervio Ciático/lesiones
14.
J Biol Chem ; 290(25): 15512-15525, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25944905

RESUMEN

Kinesin-1 is a molecular motor responsible for cargo transport along microtubules and plays critical roles in polarized cells, such as neurons. Kinesin-1 can function as a dimer of two kinesin heavy chains (KHC), which harbor the motor domain, or as a tetramer in combination with two accessory light chains (KLC). To ensure proper cargo distribution, kinesin-1 activity is precisely regulated. Both KLC and KHC subunits bind cargoes or regulatory proteins to engage the motor for movement along microtubules. We previously showed that the scaffolding protein JIP3 interacts directly with KHC in addition to its interaction with KLC and positively regulates dimeric KHC motility. Here we determined the stoichiometry of JIP3-KHC complexes and observed approximately four JIP3 molecules binding per KHC dimer. We then determined whether JIP3 activates tetrameric kinesin-1 motility. Using an in vitro motility assay, we show that JIP3 binding to KLC engages kinesin-1 with microtubules and that JIP3 binding to KHC promotes kinesin-1 motility along microtubules. We tested the in vivo relevance of these findings using axon elongation as a model for kinesin-1-dependent cellular function. We demonstrate that JIP3 binding to KHC, but not KLC, is essential for axon elongation in hippocampal neurons as well as axon regeneration in sensory neurons. These findings reveal that JIP3 regulation of kinesin-1 motility is critical for axon elongation and regeneration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Axones/metabolismo , Hipocampo/metabolismo , Cinesinas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Receptoras Sensoriales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células HEK293 , Hipocampo/citología , Humanos , Cinesinas/genética , Ratones , Microtúbulos/genética , Microtúbulos/metabolismo , Complejos Multiproteicos/genética , Proteínas del Tejido Nervioso/genética , Multimerización de Proteína/fisiología , Transporte de Proteínas/fisiología , Células Receptoras Sensoriales/citología
15.
J Biol Chem ; 290(37): 22759-70, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26157139

RESUMEN

Microtubule dynamics are important for axon growth during development as well as axon regeneration after injury. We have previously identified HDAC5 as an injury-regulated tubulin deacetylase that functions at the injury site to promote axon regeneration. However, the mechanisms involved in the spatial control of HDAC5 activity remain poorly understood. Here we reveal that HDAC5 interacts with the actin binding protein filamin A via its C-terminal domain. Filamin A plays critical roles in HDAC5-dependent tubulin deacetylation because, in cells lacking filamin A, the levels of acetylated tubulin are elevated markedly. We found that nerve injury increases filamin A axonal expression in a protein synthesis-dependent manner. Reducing filamin A levels or interfering with the interaction between HDAC5 and filamin A prevents injury-induced tubulin deacetylation as well as HDAC5 localization at the injured axon tips. In addition, neurons lacking filamin A display reduced axon regeneration. Our findings suggest a model in which filamin A local translation following axon injury controls localized HDAC5 activity to promote axon regeneration.


Asunto(s)
Axones/fisiología , Filaminas/metabolismo , Histona Desacetilasas/metabolismo , Modelos Neurológicos , Regeneración , Acetilación , Animales , Células Cultivadas , Filaminas/genética , Histona Desacetilasas/genética , Ratones , Ratones Noqueados , Transporte de Proteínas/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
16.
J Biol Chem ; 290(23): 14765-75, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25911101

RESUMEN

Injured peripheral neurons successfully activate a pro-regenerative program to enable axon regeneration and functional recovery. The microtubule-dependent retrograde transport of injury signals from the lesion site in the axon back to the cell soma stimulates the increased growth capacity of injured neurons. However, the mechanisms initiating this retrograde transport remain poorly understood. Here we show that tubulin-tyrosine ligase (TTL) is required to increase the levels of tyrosinated α-tubulin at the axon injury site and plays an important role in injury signaling. Preventing the injury-induced increase in tyrosinated α-tubulin by knocking down TTL impairs retrograde organelle transport and delays activation of the pro-regenerative transcription factor c-Jun. In the absence of TTL, axon regeneration is reduced severely. We propose a model in which TTL increases the levels of tyrosinated α-tubulin locally at the injury site to facilitate the retrograde transport of injury signals that are required to activate a pro-regenerative program.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa , Péptido Sintasas/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/fisiología , Tubulina (Proteína)/metabolismo , Animales , Axones/patología , Ratones , Nervio Ciático/patología , Tubulina (Proteína)/química , Tirosina/análisis , Tirosina/metabolismo
17.
EMBO J ; 31(14): 3063-78, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22692128

RESUMEN

Axon regeneration is an essential process to rebuild functional connections between injured neurons and their targets. Regenerative axonal growth requires alterations in axonal microtubule dynamics, but the signalling mechanisms involved remain incompletely understood. Our results reveal that axon injury induces a gradient of tubulin deacetylation, which is required for axon regeneration both in vitro and in vivo. This injury-induced tubulin deacetylation is specific to peripheral neurons and fails to occur in central neurons. We found that tubulin deacetylation is initiated by calcium influx at the site of injury, and requires protein kinase C-mediated activation of the histone deacetylase 5 (HDAC5). Our findings identify HDAC5 as a novel injury-regulated tubulin deacetylase that plays an essential role in growth cone dynamics and axon regeneration. In addition, our results suggest a mechanism for the spatial control of tubulin modifications that is required for axon regeneration.


Asunto(s)
Axones/metabolismo , Señalización del Calcio , Histona Desacetilasas/metabolismo , Regeneración Nerviosa , Proteína Quinasa C/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación , Animales , Axones/patología , Ratones
18.
BMC Vet Res ; 12: 77, 2016 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-27170186

RESUMEN

BACKGROUND: This study was aimed at evaluating the clinical protection, the level of Porcine circovirus type 2 (PCV2) viremia and the immune response (antibodies and IFN-γ secreting cells (SC)) in piglets derived from PCV2 vaccinated sows and themselves vaccinated against PCV2 at different age, namely at 4, 6 and 8 weeks. The cohort study has been carried out over three subsequent production cycles (replicates). At the start/enrolment, 46 gilts were considered at first mating, bled and vaccinated. At the first, second and third farrowing, dams were bled and re-vaccinated at the subsequent mating after weaning piglets. Overall 400 piglets at each farrowing (first, second and third) were randomly allocated in three different groups (100 piglets/group) based on the timing of vaccination (4, 6 or 8 weeks of age). A fourth group was kept non-vaccinated (controls). Piglets were vaccinated intramuscularly with one dose (2 mL) of a commercial PCV2a-based subunit vaccine (Porcilis® PCV). Twenty animals per group were bled at weaning and from vaccination to slaughter every 4 weeks for the detection of PCV2 viremia, humoral and cell-mediated immune responses. Clinical signs and individual treatments (morbidity), mortality, and body weight of all piglets were recorded. RESULTS: All vaccination schemes (4, 6 and 8 weeks of age) were able to induce an antibody response and IFN-γ SC. The highest clinical and virological protection sustained by immune reactivity was observed in pigs vaccinated at 6 weeks of age. Overall, repeated PCV2 vaccination in sows at mating and the subsequent higher levels of maternally derived antibodies did not significantly interfere with the induction of both humoral and cell-mediated immunity in their piglets after vaccination. CONCLUSIONS: The combination of vaccination in sows at mating and in piglets at 6 weeks of age was more effective for controlling PCV2 natural infection, than other vaccination schemas, thus sustaining that some interference of MDA with the induction of an efficient immune response could be considered. In conclusion, optimal vaccination strategy needs to balance the levels of passive immunity, the management practices and timing of infection.


Asunto(s)
Infecciones por Circoviridae/veterinaria , Circovirus/inmunología , Inmunidad Materno-Adquirida , Enfermedades de los Porcinos/inmunología , Vacunas Virales/inmunología , Envejecimiento/inmunología , Animales , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/inmunología , Peso Corporal , Infecciones por Circoviridae/inmunología , Infecciones por Circoviridae/prevención & control , Método Doble Ciego , Femenino , Inmunidad Celular , Interferón gamma/metabolismo , Masculino , Porcinos , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología
19.
J Biol Chem ; 289(22): 15820-32, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24737317

RESUMEN

Injured peripheral neurons successfully activate intrinsic signaling pathways to enable axon regeneration. We have previously shown that dorsal root ganglia (DRG) neurons activate the mammalian target of rapamycin (mTOR) pathway following injury and that this activity enhances their axon growth capacity. mTOR plays a critical role in protein synthesis, but the mTOR-dependent proteins enhancing the regenerative capacity of DRG neurons remain unknown. To identify proteins whose expression is regulated by injury in an mTOR-dependent manner, we analyzed the protein composition of DRGs from mice in which we genetically activated mTOR and from mice with or without a prior nerve injury. Quantitative label-free mass spectrometry analyses revealed that the injury effects were correlated with mTOR activation. We identified a member of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family of proteins, syntaxin13, whose expression was increased by injury in an mTOR-dependent manner. Increased syntaxin13 levels in injured nerves resulted from local protein synthesis and not axonal transport. Finally, knockdown of syntaxin13 in cultured DRG neurons prevented axon growth and regeneration. Together, these data suggest that syntaxin13 translation is regulated by mTOR in injured neurons to promote axon regeneration.


Asunto(s)
Regeneración Nerviosa/fisiología , Proteínas Qa-SNARE/metabolismo , Células Receptoras Sensoriales/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Axones/metabolismo , Axones/patología , Axotomía , Células Cultivadas , Femenino , Ganglios Espinales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteómica , Proteínas Qa-SNARE/genética , Nervio Ciático/metabolismo , Nervio Ciático/patología , Células Receptoras Sensoriales/patología , Serina-Treonina Quinasas TOR/genética
20.
EMBO J ; 30(16): 3416-29, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21750526

RESUMEN

Neuronal development, function and repair critically depend on axonal transport of vesicles and protein complexes, which is mediated in part by the molecular motor kinesin-1. Adaptor proteins recruit kinesin-1 to vesicles via direct association with kinesin heavy chain (KHC), the force-generating component, or via the accessory light chain (KLC). Binding of adaptors to the motor is believed to engage the motor for microtubule-based transport. We report that the adaptor protein Sunday Driver (syd, also known as JIP3 or JSAP1) interacts directly with KHC, in addition to and independently of its known interaction with KLC. Using an in vitro motility assay, we show that syd activates KHC for transport and enhances its motility, increasing both KHC velocity and run length. syd binding to KHC is functional in neurons, as syd mutants that bind KHC but not KLC are transported to axons and dendrites similarly to wild-type syd. This transport does not rely on syd oligomerization with itself or other JIP family members. These results establish syd as a positive regulator of kinesin activity and motility.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Transporte Axonal/fisiología , Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/fisiología , Animales , Células COS , Chlorocebus aethiops , Cinesinas , Ratones , Microtúbulos/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA