Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros











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

RESUMEN

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

2.
Brain ; 147(4): 1457-1473, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38066620

RESUMEN

Acyl-CoA binding domain containing 5 (ACBD5) is a critical player in handling very long chain fatty acids (VLCFA) en route for peroxisomal ß-oxidation. Mutations in ACBD5 lead to the accumulation of VLCFA and patients present retinal dystrophy, ataxia, psychomotor delay and a severe leukodystrophy. Using CRISPR/Cas9, we generated and characterized an Acbd5 Gly357* mutant allele. Gly357* mutant mice recapitulated key features of the human disorder, including reduced survival, impaired locomotion and reflexes, loss of photoreceptors, and demyelination. The ataxic presentation of Gly357* mice involved the loss of cerebellar Purkinje cells and a giant axonopathy throughout the CNS. Lipidomic studies provided evidence for the extensive lipid dysregulation caused by VLCFA accumulation. Following a proteomic survey, functional studies in neurons treated with VLCFA unravelled a deregulated cytoskeleton with reduced actin dynamics and increased neuronal filopodia. We also show that an adeno-associated virus-mediated gene delivery ameliorated the gait phenotypes and the giant axonopathy, also improving myelination and astrocyte reactivity. Collectively, we established a mouse model with significance for VLCFA-related disorders. The development of relevant neuropathological outcomes enabled the understanding of mechanisms modulated by VLCFA and the evaluation of the efficacy of preclinical therapeutic interventions.


Asunto(s)
Adrenoleucodistrofia , Ácidos Grasos , Humanos , Ratones , Animales , Ácidos Grasos/metabolismo , Dependovirus/genética , Proteómica , Ataxia , Terapia Genética , Adrenoleucodistrofia/genética
3.
Front Mol Neurosci ; 16: 1231659, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37588057

RESUMEN

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

4.
PLoS Biol ; 20(7): e3001706, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35793314

RESUMEN

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


Asunto(s)
Galactosilceramidasa , Leucodistrofia de Células Globoides , Animales , Modelos Animales de Enfermedad , Galactosilceramidasa/genética , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/patología , Ratones , Neuroglía/patología , Neuronas/fisiología
5.
Genet Med ; 24(2): 319-331, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906466

RESUMEN

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


Asunto(s)
Hidrocefalia , Discapacidad Intelectual , Agenesia del Cuerpo Calloso/genética , Agenesia del Cuerpo Calloso/patología , Animales , Variaciones en el Número de Copia de ADN , Humanos , Hidrocefalia/genética , Discapacidad Intelectual/genética , Ratones , Fenotipo
6.
Prog Neurobiol ; 205: 102123, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34302896

RESUMEN

The axon initial segment (AIS) is a specialized region in neurons that encompasses two essential functions, the generation of action potentials and the regulation of the axodendritic polarity. The mechanism controlling the position of the axon initial segment to allow plasticity and regulation of neuron excitability is unclear. Here we demonstrate that plasmalogens, the most abundant ether-phospholipid, are essential for the homeostatic positioning of the AIS. Plasmalogen deficiency is a hallmark of Rhizomelic Chondrodysplasia Punctata (RCDP) and Zellweger spectrum disorders, but Alzheimer's and Parkinson's disease, are also characterized by plasmalogen defects. Neurons lacking plasmalogens displaced the AIS to more distal positions and were characterized by reduced excitability. Treatment with a short-chain alkyl glycerol was able to rescue AIS positioning. Plasmalogen deficiency impaired AKT activation, and we show that inhibition of AKT phosphorylation at Ser473 and Thr308 is sufficient to induce a distal relocation of the AIS. Pathway analysis revealed that downstream of AKT, overtly active ULK1 mediates AIS repositioning. Rescuing the impaired AKT signaling pathway was able to normalize AIS position independently of the biochemical defect. These results unveil a previously unknown mechanism that couples the phospholipid composition of the neuronal membrane to the positional assembly of the AIS.


Asunto(s)
Segmento Inicial del Axón , Homólogo de la Proteína 1 Relacionada con la Autofagia , Condrodisplasia Punctata Rizomélica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Plasmalógenos , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal
7.
J Clin Invest ; 130(4): 2024-2040, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31945017

RESUMEN

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


Asunto(s)
Citoesqueleto , Terapia Genética , Conos de Crecimiento/metabolismo , Regeneración Nerviosa , Nervio Ciático/fisiología , Traumatismos de la Médula Espinal , Animales , Citoesqueleto/genética , Citoesqueleto/metabolismo , Dependovirus , Ratones , Ratones Noqueados , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Profilinas/biosíntesis , Profilinas/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia , Transducción Genética
8.
Brain Pathol ; 29(5): 622-639, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30667116

RESUMEN

Plasmalogens are the most abundant form of ether phospholipids in myelin and their deficiency causes Rhizomelic Chondrodysplasia Punctata (RCDP), a severe developmental disorder. Using the Gnpat-knockout (KO) mouse as a model of RCDP, we determined the consequences of a plasmalogen deficiency during myelination and myelin homeostasis in the central nervous system (CNS). We unraveled that the lack of plasmalogens causes a generalized hypomyelination in several CNS regions including the optic nerve, corpus callosum and spinal cord. The defect in myelin content evolved to a progressive demyelination concomitant with generalized astrocytosis and white matter-selective microgliosis. Oligodendrocyte precursor cells (OPC) and mature oligodendrocytes were abundant in the CNS of Gnpat KO mice during the active period of demyelination. Axonal loss was minimal in plasmalogen-deficient mice, although axonal damage was observed in spinal cords from aged Gnpat KO mice. Characterization of the plasmalogen-deficient myelin identified myelin basic protein and septin 7 as early markers of dysmyelination, whereas myelin-associated glycoprotein was associated with the active demyelination phase. Using in vitro myelination assays, we unraveled that the intrinsic capacity of oligodendrocytes to ensheath and initiate membrane wrapping requires plasmalogens. The defect in plasmalogens was rescued with glyceryl 1-myristyl ether [1-O-tetradecyl glycerol (1-O-TDG)], a novel alternative precursor in the plasmalogen biosynthesis pathway. 1-O-TDG treatment rescued myelination in plasmalogen-deficient oligodendrocytes and in mutant mice. Our results demonstrate the importance of plasmalogens for oligodendrocyte function and myelin assembly, and identified a novel strategy to promote myelination in nervous tissue.


Asunto(s)
Éteres de Glicerilo/farmacología , Oligodendroglía/metabolismo , Plasmalógenos/metabolismo , Animales , Axones/metabolismo , Sistema Nervioso Central/metabolismo , Condrodisplasia Punctata Rizomélica/metabolismo , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Leucodistrofia Metacromática/fisiopatología , Ratones , Ratones Noqueados , Vaina de Mielina/metabolismo , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Peroxisomas , Médula Espinal/metabolismo
9.
Brain Pathol ; 28(5): 631-643, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29341299

RESUMEN

Peroxisomes play a crucial role in normal neurodevelopment and in the maintenance of the adult brain. This depends largely on intact peroxisomal ß-oxidation given the similarities in pathologies between peroxisome biogenesis disorders and deficiency of multifunctional protein-2 (MFP2), the central enzyme of this pathway. Recently, adult patients diagnosed with cerebellar ataxia were shown to have mild mutations in the MFP2 gene, hydroxy-steroid dehydrogenase (17 beta) type 4 (HSD17B4). Cerebellar atrophy also develops in MFP2 deficient mice but the cellular origin of the degeneration is unexplored. In order to investigate whether peroxisomal ß-oxidation is essential within Purkinje cells, the sole output neurons of the cerebellum, we generated and characterized a mouse model with Purkinje cell selective deletion of the MFP2 gene. We show that selective loss of MFP2 from mature cerebellar Purkinje neurons causes a late-onset motor phenotype and progressive Purkinje cell degeneration, thereby mimicking ataxia and cerebellar deterioration in patients with mild HSD17B4 mutations. We demonstrate that swellings on Purkinje cell axons coincide with ataxic behavior and precede neurodegeneration. Loss of Purkinje cells occurs in a characteristic banded pattern, proceeds in an anterior to posterior fashion and is accompanied by progressive astro- and microgliosis. These data prove that the peroxisomal ß-oxidation pathway is required within Purkinje neurons to maintain their axonal integrity, independent of glial dysfunction.


Asunto(s)
Axones/fisiología , Ataxia Cerebelosa/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Trastorno Peroxisomal/fisiopatología , Proteína-2 Multifuncional Peroxisomal/deficiencia , Células de Purkinje/fisiología , Envejecimiento , Animales , Astrocitos/patología , Astrocitos/fisiología , Axones/patología , Ataxia Cerebelosa/etiología , Ataxia Cerebelosa/patología , Modelos Animales de Enfermedad , Gliosis/patología , Gliosis/fisiopatología , Ratones Transgénicos , Microglía/patología , Microglía/fisiología , Enfermedades Neurodegenerativas/patología , Trastorno Peroxisomal/patología , Proteína-2 Multifuncional Peroxisomal/genética , Células de Purkinje/patología
10.
Mitochondrion ; 39: 51-59, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28866057

RESUMEN

The structural disruption of the mitochondrial inner membrane in hepatocytes lacking functional peroxisomes along with selective impairment of respiratory complexes and depletion of mitochondrial DNA was previously reported. In search for the molecular origin of these mitochondrial alterations, we here show that these are tissue selective as they do neither occur in peroxisome deficient brain nor in peroxisome deficient striated muscle. Given the hepatocyte selectivity, we investigated the potential involvement of metabolites that are primarily handled by hepatic peroxisomes. Levels of these metabolites were manipulated in L-Pex5 knockout mice and/or compared with levels in different mouse models with a peroxisomal ß-oxidation deficiency. We show that neither the deficiency of docosahexaenoic acid nor the accumulation of branched chain fatty acids, dicarboxylic acids or C27 bile acid intermediates are solely responsible for the mitochondrial anomalies. In conclusion, we demonstrate that peroxisomal inactivity differentially impacts mitochondria depending on the cell type but the cause of the mitochondrial destruction needs to be further explored.


Asunto(s)
Hepatocitos/enzimología , Hepatocitos/patología , Hígado/enzimología , Hígado/patología , Mitocondrias/patología , Peroxisomas/patología , Animales , Encéfalo/enzimología , Encéfalo/patología , ADN Mitocondrial/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/deficiencia , Ratones Noqueados , Membranas Mitocondriales/patología , Músculo Estriado/enzimología , Músculo Estriado/patología
11.
Cereb Cortex ; 27(3): 1732-1747, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334068

RESUMEN

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


Asunto(s)
Axones/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proyección Neuronal , Proteína Smad2/metabolismo , Envejecimiento/metabolismo , Animales , Aumento de la Célula , Línea Celular , Células Cultivadas , Femenino , Ganglios Espinales/metabolismo , Hipocampo/metabolismo , Humanos , Janus Quinasa 2/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Dominios Proteicos , Ratas Wistar , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Médula Espinal/metabolismo
12.
J Neurosci Res ; 94(11): 1037-41, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27638589

RESUMEN

In Krabbe's disease (KD), demyelination and myelin-independent axonal and neuronal defects contribute to the severe neuropathology. The toxic substrate that accumulates in this disease, psychosine, induces alterations in membrane lipid rafts with downstream consequences to cellular signaling pathways that include impaired protein kinase C, ERK, and AKT-glycogen synthase kinase-3ß (GSK3ß) activation. In addition to impaired recruitment of signaling proteins to lipid rafts, endocytosis and axonal transport are affected in KD. Defects in AKT-GSK3ß activation, a central pathway regulating microtubule stability, together with alterations in neurofilaments and microtubules and severely defective axonal transport, highlight the importance of the neuronal cytoskeleton in KD. This Review critically discusses these primary neuronal defects as well as new windows for action opened by their identification that may contribute to effectively correct the neuropathology that underlies this disorder. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Axones/patología , Citoesqueleto/fisiología , Leucodistrofia de Células Globoides/patología , Leucodistrofia de Células Globoides/terapia , Neuronas/patología , Axones/metabolismo , Psicosina/toxicidad
13.
Neurobiol Dis ; 94: 157-68, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27353294

RESUMEN

The cerebellar pathologies in peroxisomal diseases underscore that these organelles are required for the normal development and maintenance of the cerebellum, but the mechanisms have not been resolved. Here we investigated the origins of the early-onset coordination impairment in a mouse model with neural selective deficiency of multifunctional protein-2, the central enzyme of peroxisomal ß-oxidation. At the age of 4weeks, Nestin-Mfp2(-/-) mice showed impaired motor learning on the accelerating rotarod and underperformed on the balance beam test. The gross morphology of the cerebellum and Purkinje cell arborization were normal. However, electrophysiology revealed a reduced Purkinje cell firing rate, a decreased excitability and an increased membrane capacitance. The distribution of climbing and parallel fiber synapses on Purkinje cells was immature and was accompanied by an increased spine length. Despite normal myelination, Purkinje cell axon degeneration was evident from the occurrence of axonal swellings containing accumulated organelles. In conclusion, the electrical activity, axonal integrity and wiring of Purkinje cells are exquisitely dependent on intact peroxisomal ß-oxidation in neural cells.


Asunto(s)
Cerebelo/metabolismo , Proteína-2 Multifuncional Peroxisomal/metabolismo , Células de Purkinje/metabolismo , Sinapsis/fisiología , Animales , Axones/metabolismo , Ataxia Cerebelosa/metabolismo , Ratones Noqueados , Proteína-2 Multifuncional Peroxisomal/deficiencia
14.
Cell Rep ; 15(3): 490-498, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27068466

RESUMEN

The actin-binding protein adducin was recently identified as a component of the neuronal subcortical cytoskeleton. Here, we analyzed mice lacking adducin to uncover the function of this protein in actin rings. α-adducin knockout mice presented progressive axon enlargement in the spinal cord and optic and sciatic nerves, followed by axon degeneration and loss. Using stimulated emission depletion super-resolution microscopy, we show that a periodic subcortical actin cytoskeleton is assembled in every neuron type inspected including retinal ganglion cells and dorsal root ganglia neurons. In neurons devoid of adducin, the actin ring diameter increased, although the inter-ring periodicity was maintained. In vitro, the actin ring diameter adjusted as axons grew, suggesting the lattice is dynamic. Our data support a model in which adducin activity is not essential for actin ring assembly and periodicity but is necessary to control the diameter of both actin rings and axons and actin filament growth within rings.


Asunto(s)
Axones/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Proteínas de Microfilamentos/metabolismo , Animales , Transporte Axonal , Citoesqueleto/metabolismo , Conos de Crecimiento/metabolismo , Hipocampo/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología
15.
Mol Neurobiol ; 53(2): 1052-1064, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25579385

RESUMEN

Lack of axon regeneration following spinal cord injury has been mainly ascribed to the inhibitory environment of the injury site, i.e., to chondroitin sulfate proteoglycans (CSPGs) and myelin-associated inhibitors (MAIs). Here, we used shiverer (shi) mice to assess axon regeneration following spinal cord injury in the presence of MAIs and CSPG but in the absence of compact myelin. Although in vitro shi neurons displayed a similar intrinsic neurite outgrowth to wild-type neurons, in vivo, shi fibers had increased regenerative capacity, suggesting that the wild-type spinal cord contains additional inhibitors besides MAIs and CSPG. Our data show that besides myelin protein, myelin lipids are highly inhibitory for neurite outgrowth and suggest that this inhibitory effect is released in the shi spinal cord given its decreased lipid content. Specifically, we identified cholesterol and sphingomyelin as novel myelin-associated inhibitors that operate through a Rho-dependent mechanism and have inhibitory activity in multiple neuron types. We further demonstrated the inhibitory action of myelin lipids in vivo, by showing that delivery of 2-hydroxypropyl-ß-cyclodextrin, a drug that reduces the levels of lipids specifically in the injury site, leads to increased axon regeneration of wild-type (WT) dorsal column axons following spinal cord injury. In summary, our work shows that myelin lipids are important modulators of axon regeneration that should be considered together with protein MAIs as critical targets in strategies aiming at improving axonal growth following injury.


Asunto(s)
Axones/patología , Lípidos/química , Vaina de Mielina/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia , Médula Espinal/patología , 2-Hidroxipropil-beta-Ciclodextrina , Animales , Colesterol/metabolismo , Ratones Endogámicos C57BL , Vaina de Mielina/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/patología , Esfingomielinas/metabolismo , Médula Espinal/efectos de los fármacos , beta-Ciclodextrinas/farmacología , Proteínas de Unión al GTP rho/metabolismo
16.
Free Radic Biol Med ; 84: 296-310, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25801291

RESUMEN

Reactive oxygen species (ROS) are implicated in a range of degenerative conditions, including aging, neurodegenerative diseases, and neurological disorders. Myelin is a lipid-rich multilamellar sheath that facilitates rapid nerve conduction in vertebrates. Given the high energetic demands and low antioxidant capacity of the cells that elaborate the sheaths, myelin is considered intrinsically vulnerable to oxidative damage, raising the question whether additional mechanisms prevent structural damage. We characterized the structural and biochemical basis of ROS-mediated myelin damage in murine tissues from both central nervous system (CNS) and peripheral nervous system (PNS). To determine whether ROS can cause structural damage to the internodal myelin, whole sciatic and optic nerves were incubated ex vivo with a hydroxyl radical-generating system consisting of copper (Cu), hydrogen peroxide (HP), and ortho-phenanthroline (OP). Quantitative assessment of unfixed tissue by X-ray diffraction revealed irreversible compaction of myelin membrane stacking in both sciatic and optic nerves. Incubation in the presence of the hydroxyl radical scavenger sodium formate prevented this damage, implicating hydroxyl radical species. Myelin membranes are particularly enriched in plasmalogens, a class of ether-linked phospholipids proposed to have antioxidant properties. Myelin in sciatic nerve from plasmalogen-deficient (Pex7 knockout) mice was significantly more vulnerable to Cu/OP/HP-mediated ROS-induced compaction than myelin from WT mice. Our results directly support the role of plasmalogens as endogenous antioxidants providing a defense that protects ROS-vulnerable myelin.


Asunto(s)
Depuradores de Radicales Libres/farmacología , Vaina de Mielina/metabolismo , Plasmalógenos/farmacología , Animales , Quelantes/farmacología , Evaluación Preclínica de Medicamentos , Ácido Edético/farmacología , Formiatos/farmacología , Ratones Noqueados , Vaina de Mielina/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/patología , Oxidación-Reducción , Estrés Oxidativo , Receptor de la Señal 2 de Direccionamiento al Peroxisoma , Carbonilación Proteica , Especies Reactivas de Oxígeno/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Nervio Ciático/metabolismo , Nervio Ciático/patología
17.
J Inherit Metab Dis ; 38(1): 111-21, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25432520

RESUMEN

Plasmalogens are a special class of ether-phospholipids, best recognized by their vinyl-ether bond at the sn-1 position of the glycerobackbone and by the observation that their deficiency causes rhizomelic chondrodysplasia punctata (RCDP). The complex plasmalogen biosynthetic pathway involves multiple enzymatic steps carried-out in peroxisomes and in the endoplasmic reticulum. The rate limiting step in the biosynthesis of plasmalogens resides in the formation of the fatty alcohol responsible for the formation of an intermediate with an alkyl-linked moiety. The regulation in the biosynthesis of plasmalogens also takes place at this step using a feedback mechanism to stimulate or inhibit the biosynthesis. As such, fatty alcohols play a relevant role in the formation of ether-phospholipids. These advances in our understanding of complex lipid biosynthesis brought two seemingly distinct disorders into the spotlight. Sjögren-Larsson syndrome (SLS) is caused by defects in the microsomal fatty aldehyde dehydrogenase (FALDH) leading to the accumulation of fatty alcohols and fatty aldehydes. In RCDP cells, the defect in plasmalogens is thought to generate a feedback signal to increase their biosynthesis, through the activity of fatty acid reductases to produce fatty alcohols. However, the enzymatic defects in either glyceronephosphate O-acyltransferase (GNPAT) or alkylglycerone phosphate synthase (AGPS) disrupt the biosynthesis and result in the accumulation of the fatty alcohols. A detailed characterization on the processes and enzymes that govern these intricate biosynthetic pathways, as well as, the metabolic characterization of defects along the pathway should increase our understanding of the causes and mechanisms behind these disorders.


Asunto(s)
Condrodisplasia Punctata Rizomélica/metabolismo , Alcoholes Grasos/metabolismo , Plasmalógenos/metabolismo , Síndrome de Sjögren-Larsson/metabolismo , Aldehído Oxidorreductasas/metabolismo , Aldehídos/metabolismo , Animales , Antioxidantes/metabolismo , Ácidos Grasos/metabolismo , Humanos , Ratones , Microsomas/metabolismo , Peroxisomas/metabolismo
18.
Mol Cell Biol ; 34(15): 2917-28, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24865970

RESUMEN

Peroxisomal matrix proteins are synthesized on cytosolic ribosomes and transported to the organelle by shuttling receptors. Matrix proteins containing a type 1 signal are carried to the peroxisome by PEX5, whereas those harboring a type 2 signal are transported by a PEX5-PEX7 complex. The pathway followed by PEX5 during the protein transport cycle has been characterized in detail. In contrast, not much is known regarding PEX7. In this work, we show that PEX7 is targeted to the peroxisome in a PEX5- and cargo-dependent manner, where it becomes resistant to exogenously added proteases. Entry of PEX7 and its cargo into the peroxisome occurs upstream of the first cytosolic ATP-dependent step of the PEX5-mediated import pathway, i.e., before monoubiquitination of PEX5. PEX7 passing through the peroxisome becomes partially, if not completely, exposed to the peroxisome matrix milieu, suggesting that cargo release occurs at the trans side of the peroxisomal membrane. Finally, we found that export of peroxisomal PEX7 back into the cytosol requires export of PEX5 but, strikingly, the two export events are not strictly coupled, indicating that the two proteins leave the peroxisome separately.


Asunto(s)
Peroxisomas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Proteínas Portadoras/metabolismo , Humanos , Membranas/metabolismo , Ratones , Receptor de la Señal 2 de Direccionamiento al Peroxisoma , Transporte de Proteínas/fisiología , Conejos , Ratas , Transducción de Señal/fisiología
19.
J Clin Invest ; 124(6): 2560-70, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24762439

RESUMEN

Rhizomelic chondrodysplasia punctata (RCDP) is a developmental disorder characterized by hypotonia, cataracts, abnormal ossification, impaired motor development, and intellectual disability. The underlying etiology of RCDP is a deficiency in the biosynthesis of ether phospholipids, of which plasmalogens are the most abundant form in nervous tissue and myelin; however, the role of plasmalogens in the peripheral nervous system is poorly defined. Here, we used mouse models of RCDP and analyzed the consequence of plasmalogen deficiency in peripheral nerves. We determined that plasmalogens are crucial for Schwann cell development and differentiation and that plasmalogen defects impaired radial sorting, myelination, and myelin structure. Plasmalogen insufficiency resulted in defective protein kinase B (AKT) phosphorylation and subsequent signaling, causing overt activation of glycogen synthase kinase 3ß (GSK3ß) in nerves of mutant mice. Treatment with GSK3ß inhibitors, lithium, or 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) restored Schwann cell defects, effectively bypassing plasmalogen deficiency. Our results demonstrate the requirement of plasmalogens for the correct and timely differentiation of Schwann cells and for the process of myelination. In addition, these studies identify a mechanism by which the lack of a membrane phospholipid causes neuropathology, implicating plasmalogens as regulators of membrane and cell signaling.


Asunto(s)
Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/fisiología , Plasmalógenos/fisiología , Células de Schwann/citología , Células de Schwann/fisiología , Animales , Diferenciación Celular/fisiología , Condrodisplasia Punctata Rizomélica/etiología , Condrodisplasia Punctata Rizomélica/patología , Condrodisplasia Punctata Rizomélica/fisiopatología , Femenino , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Modelos Neurológicos , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/fisiología , Regeneración Nerviosa , Receptor de la Señal 2 de Direccionamiento al Peroxisoma , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
20.
PLoS One ; 9(2): e88593, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586346

RESUMEN

Mammalian central nervous system (CNS) neurons do not regenerate after injury due to the inhibitory environment formed by the glial scar, largely constituted by myelin debris. The use of biomaterials to bridge the lesion area and the creation of an environment favoring axonal regeneration is an appealing approach, currently under investigation. This work aimed at assessing the suitability of three candidate polymers - poly(ε-caprolactone), poly(trimethylene carbonate-co-ε-caprolactone) (P(TMC-CL)) (11∶89 mol%) and poly(trimethylene carbonate) - with the final goal of using these materials in the development of conduits to promote spinal cord regeneration. Poly(L-lysine) (PLL) coated polymeric films were tested for neuronal cell adhesion and neurite outgrowth. At similar PLL film area coverage conditions, neuronal polarization and axonal elongation was significantly higher on P(TMC-CL) films. Furthermore, cortical neurons cultured on P(TMC-CL) were able to extend neurites even when seeded onto myelin. This effect was found to be mediated by the glycogen synthase kinase 3ß (GSK3ß) signaling pathway with impact on the collapsin response mediator protein 4 (CRMP4), suggesting that besides surface topography, nanomechanical properties were implicated in this process. The obtained results indicate P(TMC-CL) as a promising material for CNS regenerative applications as it promotes axonal growth, overcoming myelin inhibition.


Asunto(s)
Axones/efectos de los fármacos , Axones/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Poliésteres/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Vaina de Mielina/metabolismo , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA