Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Cell Rep ; 40(13): 111413, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36170840

RESUMEN

Efficient myelination supports nerve conduction and axonal health throughout life. In the central nervous system, oligodendrocytes (OLs) carry out this demanding anabolic duty in part through biosynthetic pathways controlled by mTOR. We identify Ral GTPases as critical regulators of mouse spinal cord myelination and myelin maintenance. Ablation of Ral GTPases (RalA, RalB) in OL-lineage cells impairs timely onset and radial growth of developmental myelination, accompanied by increased endosomal/lysosomal abundance. Further examinations, including transcriptomic analyses of Ral-deficient OLs, were consistent with mTORC1-related deficits. However, deletion of the mTOR signaling-repressor Pten in Ral-deficient OL-lineage cells is unable to rescue mTORC1 activation or developmental myelination deficiencies. Induced deletion of Ral GTPases in OLs of adult mice results in late-onset myelination defects and tissue degeneration. Together, our data indicate critical roles for Ral GTPases to promote developmental spinal cord myelination, to ensure accurate mTORC1 signaling, and to protect the healthy state of myelin-axon units over time.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Proteínas de Unión al GTP ral , Animales , Homeostasis , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Proteínas de Unión al GTP Monoméricas/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión al GTP ral/metabolismo
2.
Elife ; 102021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33890853

RESUMEN

Peripheral nerves are organ-like structures containing diverse cell types to optimize function. This interactive assembly includes mostly axon-associated Schwann cells, but also endothelial cells of supporting blood vessels, immune system-associated cells, barrier-forming cells of the perineurium surrounding and protecting nerve fascicles, and connective tissue-resident cells within the intra-fascicular endoneurium and inter-fascicular epineurium. We have established transcriptional profiles of mouse sciatic nerve-inhabitant cells to foster the fundamental understanding of peripheral nerves. To achieve this goal, we have combined bulk RNA sequencing of developing sciatic nerves up to the adult with focused bulk and single-cell RNA sequencing of Schwann cells throughout postnatal development, extended by single-cell transcriptome analysis of the full sciatic nerve both perinatally and in the adult. The results were merged in the transcriptome resource Sciatic Nerve ATlas (SNAT: https://www.snat.ethz.ch). We anticipate that insights gained from our multi-layered analysis will serve as valuable interactive reference point to guide future studies.


Asunto(s)
Nervios Periféricos/metabolismo , Transcripción Genética , Animales , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Nervio Ciático/metabolismo
3.
Hum Mol Genet ; 29(8): 1253-1273, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32129442

RESUMEN

Some mutations affecting dynamin 2 (DNM2) can cause dominantly inherited Charcot-Marie-Tooth (CMT) neuropathy. Here, we describe the analysis of mice carrying the DNM2 K562E mutation which has been associated with dominant-intermediate CMT type B (CMTDIB). Contrary to our expectations, heterozygous DNM2 K562E mutant mice did not develop definitive signs of an axonal or demyelinating neuropathy. Rather, we found a primary myopathy-like phenotype in these mice. A likely interpretation of these results is that the lack of a neuropathy in this mouse model has allowed the unmasking of a primary myopathy due to the DNM2 K562E mutation which might be overshadowed by the neuropathy in humans. Consequently, we hypothesize that a primary myopathy may also contribute to the disease mechanism in some CMTDIB patients. We propose that these findings should be considered in the evaluation of patients, the determination of the underlying disease processes and the development of tailored potential treatment strategies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Dinamina II/deficiencia , Enfermedades Musculares/genética , Miopatías Estructurales Congénitas/genética , Animales , Axones/metabolismo , Axones/patología , Enfermedad de Charcot-Marie-Tooth/patología , Dinamina II/genética , Heterocigoto , Humanos , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/patología , Mutación/genética , Miopatías Estructurales Congénitas/patología , Fenotipo
4.
J Cell Biol ; 218(7): 2350-2369, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31201267

RESUMEN

Small GTPases of the Rho and Ras families are important regulators of Schwann cell biology. The Ras-like GTPases RalA and RalB act downstream of Ras in malignant peripheral nerve sheath tumors. However, the physiological role of Ral proteins in Schwann cell development is unknown. Using transgenic mice with ablation of one or both Ral genes, we report that Ral GTPases are crucial for axonal radial sorting. While lack of only one Ral GTPase was dispensable for early peripheral nerve development, ablation of both RalA and RalB resulted in persistent radial sorting defects, associated with hallmarks of deficits in Schwann cell process formation and maintenance. In agreement, ex vivo-cultured Ral-deficient Schwann cells were impaired in process extension and the formation of lamellipodia. Our data indicate further that RalA contributes to Schwann cell process extensions through the exocyst complex, a known effector of Ral GTPases, consistent with an exocyst-mediated function of Ral GTPases in Schwann cells.


Asunto(s)
Sistema Nervioso Periférico/crecimiento & desarrollo , Células de Schwann/metabolismo , Proteínas de Unión al GTP ral/genética , Animales , Axones/metabolismo , Movimiento Celular/genética , Células Cultivadas , Exocitosis/genética , GTP Fosfohidrolasas/genética , Humanos , Ratones , Ratones Transgénicos , Sistema Nervioso Periférico/metabolismo , Transducción de Señal/genética
5.
Elife ; 82019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31063129

RESUMEN

Oligodendrocytes (OLs) support neurons and signal transmission in the central nervous system (CNS) by enwrapping axons with myelin, a lipid-rich membrane structure. We addressed the significance of fatty acid (FA) synthesis in OLs by depleting FA synthase (FASN) from OL progenitor cells (OPCs) in transgenic mice. While we detected no effects in proliferation and differentiation along the postnatal OL lineage, we found that FASN is essential for accurate myelination, including myelin growth. Increasing dietary lipid intake could partially compensate for the FASN deficiency. Furthermore, FASN contributes to correct myelin lipid composition and stability of myelinated axons. Moreover, we depleted FASN specifically in adult OPCs to examine its relevance for remyelination. Applying lysolecithin-induced focal demyelinating spinal cord lesions, we found that FA synthesis is essential to sustain adult OPC-derived OLs and efficient remyelination. We conclude that FA synthesis in OLs plays key roles in CNS myelination and remyelination.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Ácidos Grasos/metabolismo , Vaina de Mielina/metabolismo , Células-Madre Neurales/fisiología , Oligodendroglía/metabolismo , Remielinización , Animales , Diferenciación Celular , Proliferación Celular , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Acido Graso Sintasa Tipo I/deficiencia , Acido Graso Sintasa Tipo I/metabolismo , Ratones Transgénicos
6.
Elife ; 82019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30648534

RESUMEN

Myelination requires extensive plasma membrane rearrangements, implying that molecules controlling membrane dynamics play prominent roles. The large GTPase dynamin 2 (DNM2) is a well-known regulator of membrane remodeling, membrane fission, and vesicular trafficking. Here, we genetically ablated Dnm2 in Schwann cells (SCs) and in oligodendrocytes of mice. Dnm2 deletion in developing SCs resulted in severely impaired axonal sorting and myelination onset. Induced Dnm2 deletion in adult SCs caused a rapidly-developing peripheral neuropathy with abundant demyelination. In both experimental settings, mutant SCs underwent prominent cell death, at least partially due to cytokinesis failure. Strikingly, when Dnm2 was deleted in adult SCs, non-recombined SCs still expressing DNM2 were able to remyelinate fast and efficiently, accompanied by neuropathy remission. These findings reveal a remarkable self-healing capability of peripheral nerves that are affected by SC loss. In the central nervous system, however, we found no major defects upon Dnm2 deletion in oligodendrocytes.


Asunto(s)
Dinamina II/metabolismo , Oligodendroglía/metabolismo , Células de Schwann/metabolismo , Animales , Axones/metabolismo , Muerte Celular , Diferenciación Celular , Supervivencia Celular , Citocinesis , Ratones , Mitosis , Vaina de Mielina/metabolismo , Nervios Periféricos/metabolismo , Transcriptoma/genética
7.
Glia ; 66(12): 2632-2644, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30295958

RESUMEN

Proper function of the nervous system depends on myelination. In peripheral nerves, Schwann cells (SCs) myelinate axons and the miRNA biogenesis pathway is required for developmental myelination and myelin maintenance. However, regulatory roles of this pathway at different stages of myelination are only partially understood. We addressed the requirement of the core miRNA biogenesis pathway components Dgcr8, Drosha, and Dicer in developing and adult SCs using mouse mutants with a comparative genetics and transcriptomics approach. We found that the microprocessor components Dgcr8 and Drosha are crucial for axonal radial sorting and to establish correct SC numbers upon myelination. Transcriptome analyses revealed a requirement of the microprocessor to prevent aberrantly increased expression of injury-response genes. Those genes are predicted targets of abundant miRNAs in sciatic nerves (SNs) during developmental myelination. In agreement, Dgcr8 and Dicer are required for proper maintenance of the myelinated SC state, where abundant miRNAs in adult SNs are predicted to target injury-response genes. We conclude that the miRNA biogenesis pathway in SCs is crucial for preventing inappropriate activity of injury-response genes in developing and adult SCs.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , MicroARNs/metabolismo , Células de Schwann/patología , Neuropatía Ciática/patología , Neuropatía Ciática/prevención & control , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Conexinas/genética , Conexinas/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Microscopía Electrónica , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Células de Schwann/metabolismo , Células de Schwann/ultraestructura , Factores de Transcripción/metabolismo , Proteína beta1 de Unión Comunicante
8.
Aging (Albany NY) ; 10(5): 861-862, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29788000
9.
J Neurosci ; 38(20): 4811-4828, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29695414

RESUMEN

Schwann cells (SCs) are endowed with a remarkable plasticity. When peripheral nerves are injured, SCs dedifferentiate and acquire new functions to coordinate nerve repair as so-called repair SCs. Subsequently, SCs redifferentiate to remyelinate regenerated axons. Given the similarities between SC dedifferentiation/redifferentiation in injured nerves and in demyelinating neuropathies, elucidating the signals involved in SC plasticity after nerve injury has potentially wider implications. c-Jun has emerged as a key transcription factor regulating SC dedifferentiation and the acquisition of repair SC features. However, the upstream pathways that control c-Jun activity after nerve injury are largely unknown. We report that the mTORC1 pathway is transiently but robustly reactivated in dedifferentiating SCs. By inducible genetic deletion of the functionally crucial mTORC1-subunit Raptor in mouse SCs (including male and female animals), we found that mTORC1 reactivation is necessary for proper myelin clearance, SC dedifferentiation, and consequently remyelination, without major alterations in the inflammatory response. In the absence of mTORC1 signaling, c-Jun failed to be upregulated correctly. Accordingly, a c-Jun binding motif was found to be enriched in promoters of genes with reduced expression in injured mutants. Furthermore, using cultured SCs, we found that mTORC1 is involved in c-Jun regulation by promoting its translation, possibly via the eIF4F-subunit eIF4A. These results provide evidence that proper c-Jun elevation after nerve injury involves also mTORC1-dependent post-transcriptional regulation to ensure timely dedifferentiation of SCs.SIGNIFICANCE STATEMENT A crucial evolutionary acquisition of vertebrates is the envelopment of axons in myelin sheaths produced by oligodendrocytes in the CNS and Schwann cells (SCs) in the PNS. When myelin is damaged, conduction of action potentials along axons slows down or is blocked, leading to debilitating diseases. Unlike oligodendrocytes, SCs have a high regenerative potential, granted by their remarkable plasticity. Thus, understanding the mechanisms underlying SC plasticity may uncover new therapeutic targets in nerve regeneration and demyelinating diseases. Our work reveals that reactivation of the mTORC1 pathway in SCs is essential for efficient SC dedifferentiation after nerve injury. Accordingly, modulating this signaling pathway might be of therapeutic relevance in peripheral nerve injury and other diseases.


Asunto(s)
Desdiferenciación Celular , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/metabolismo , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Células de Schwann , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Activación Metabólica/genética , Activación Metabólica/fisiología , Animales , Factor 4F Eucariótico de Iniciación/genética , Femenino , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Vaina de Mielina/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Ratas , Ratas Sprague-Dawley , Proteína Reguladora Asociada a mTOR/genética , Transducción de Señal/fisiología
10.
J Cell Biol ; 217(4): 1353-1368, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29434029

RESUMEN

Myelination calls for a remarkable surge in cell metabolism to facilitate lipid and membrane production. Endogenous fatty acid (FA) synthesis represents a potentially critical process in myelinating glia. Using genetically modified mice, we show that Schwann cell (SC) intrinsic activity of the enzyme essential for de novo FA synthesis, fatty acid synthase (FASN), is crucial for precise lipid composition of peripheral nerves and fundamental for the correct onset of myelination and proper myelin growth. Upon FASN depletion in SCs, epineurial adipocytes undergo lipolysis, suggestive of a compensatory role. Mechanistically, we found that a lack of FASN in SCs leads to an impairment of the peroxisome proliferator-activated receptor (PPAR) γ-regulated transcriptional program. In agreement, defects in myelination of FASN-deficient SCs could be ameliorated by treatment with the PPARγ agonist rosiglitazone ex vivo and in vivo. Our results reveal that FASN-driven de novo FA synthesis in SCs is mandatory for myelination and identify lipogenic activation of the PPARγ transcriptional network as a putative downstream functional mediator.


Asunto(s)
Ácidos Grasos/biosíntesis , Lipogénesis , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Células de Schwann/metabolismo , Nervio Ciático/metabolismo , Animales , Células Cultivadas , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Femenino , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Fibras Nerviosas Mielínicas/efectos de los fármacos , PPAR gamma/agonistas , PPAR gamma/metabolismo , Rosiglitazona/farmacología , Células de Schwann/efectos de los fármacos , Nervio Ciático/citología , Nervio Ciático/efectos de los fármacos , Transducción de Señal , Transcripción Genética
11.
Nat Commun ; 9(1): 236, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29339718

RESUMEN

Cutaneous wound healing is a complex process that aims to re-establish the original structure of the skin and its functions. Among other disorders, peripheral neuropathies are known to severely impair wound healing capabilities of the skin, revealing the importance of skin innervation for proper repair. Here, we report that peripheral glia are crucially involved in this process. Using a mouse model of wound healing, combined with in vivo fate mapping, we show that injury activates peripheral glia by promoting de-differentiation, cell-cycle re-entry and dissemination of the cells into the wound bed. Moreover, injury-activated glia upregulate the expression of many secreted factors previously associated with wound healing and promote myofibroblast differentiation by paracrine modulation of TGF-ß signalling. Accordingly, depletion of these cells impairs epithelial proliferation and wound closure through contraction, while their expansion promotes myofibroblast formation. Thus, injury-activated glia and/or their secretome might have therapeutic potential in human wound healing disorders.


Asunto(s)
Diferenciación Celular/fisiología , Neuroglía/fisiología , Piel/fisiopatología , Cicatrización de Heridas/fisiología , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Diferenciación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/metabolismo , Miofibroblastos/fisiología , Neuroglía/citología , Neuroglía/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Transducción de Señal/genética , Piel/lesiones , Piel/inervación , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/genética
12.
Glia ; 66(4): 693-707, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29210103

RESUMEN

Myelinating cells surround axons to accelerate the propagation of action potentials, to support axonal health, and to refine neural circuits. Myelination is metabolically demanding and, consistent with this notion, mTORC1-a signaling hub coordinating cell metabolism-has been implicated as a key signal for myelination. Here, we will discuss metabolic aspects of myelination, illustrate the main metabolic processes regulated by mTORC1, and review advances on the role of mTORC1 in myelination of the central nervous system and the peripheral nervous system. Recent progress has revealed a complex role of mTORC1 in myelinating cells that includes, besides positive regulation of myelin growth, additional critical functions in the stages preceding active myelination. Based on the available evidence, we will also highlight potential nonoverlapping roles between mTORC1 and its known main upstream pathways PI3K-Akt, Mek-Erk1/2, and AMPK in myelinating cells. Finally, we will discuss signals that are already known or hypothesized to be responsible for the regulation of mTORC1 activity in myelinating cells.


Asunto(s)
Vaina de Mielina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Humanos
13.
Elife ; 62017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28880149

RESUMEN

Myelination is a biosynthetically demanding process in which mTORC1, the gatekeeper of anabolism, occupies a privileged regulatory position. We have shown previously that loss of mTORC1 function in Schwann cells (SCs) hampers myelination. Here, we genetically disrupted key inhibitory components upstream of mTORC1, TSC1 or PTEN, in mouse SC development, adult homeostasis, and nerve injury. Surprisingly, the resulting mTORC1 hyperactivity led to markedly delayed onset of both developmental myelination and remyelination after injury. However, if mTORC1 was hyperactivated after myelination onset, radial hypermyelination was observed. At early developmental stages, physiologically high PI3K-Akt-mTORC1 signaling suppresses expression of Krox20 (Egr2), the master regulator of PNS myelination. This effect is mediated by S6K and contributes to control mechanisms that keep SCs in a not-fully differentiated state to ensure proper timing of myelination initiation. An ensuing decline in mTORC1 activity is crucial to allow myelination to start, while remaining mTORC1 activity drives myelin growth.


Asunto(s)
Vaina de Mielina/metabolismo , Sistema Nervioso Periférico/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Células Cultivadas , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/metabolismo , Sistema Nervioso Periférico/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Células de Schwann/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
14.
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
15.
Sci Rep ; 6: 36930, 2016 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-27841286

RESUMEN

Mutations in the ganglioside-induced differentiation associated protein 1 (GDAP1) cause severe peripheral motor and sensory neuropathies called Charcot-Marie-Tooth disease. GDAP1 expression induces fission of mitochondria and peroxisomes by a currently elusive mechanism, while disease causing mutations in GDAP1 impede the protein's role in mitochondrial dynamics. In silico analysis reveals sequence similarities of GDAP1 to glutathione S-transferases (GSTs). However, a proof of GST activity and its possible impact on membrane dynamics are lacking to date. Using recombinant protein, we demonstrate for the first time theta-class-like GST activity for GDAP1, and it's activity being regulated by the C-terminal hydrophobic domain 1 (HD1) of GDAP1 in an autoinhibitory manner. Moreover, we show that the HD1 amphipathic pattern is required to induce membrane dynamics by GDAP1. As both, fission and GST activities of GDAP1, are critically dependent on HD1, we propose that GDAP1 undergoes a molecular switch, turning from a pro-fission active to an auto-inhibited inactive conformation.


Asunto(s)
Membrana Celular/metabolismo , Glutatión/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Animales , Glutatión Transferasa/metabolismo , Células HEK293 , Homeostasis , Humanos , Liposomas/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Dominios Proteicos , Multimerización de Proteína , Células Sf9
16.
Sci Adv ; 2(8): e1600060, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27493992

RESUMEN

Cranial neural crest cells populate the future facial region and produce ectomesenchyme-derived tissues, such as cartilage, bone, dermis, smooth muscle, adipocytes, and many others. However, the contribution of individual neural crest cells to certain facial locations and the general spatial clonal organization of the ectomesenchyme have not been determined. We investigated how neural crest cells give rise to clonally organized ectomesenchyme and how this early ectomesenchyme behaves during the developmental processes that shape the face. Using a combination of mouse and zebrafish models, we analyzed individual migration, cell crowd movement, oriented cell division, clonal spatial overlapping, and multilineage differentiation. The early face appears to be built from multiple spatially defined overlapping ectomesenchymal clones. During early face development, these clones remain oligopotent and generate various tissues in a given location. By combining clonal analysis, computer simulations, mouse mutants, and live imaging, we show that facial shaping results from an array of local cellular activities in the ectomesenchyme. These activities mostly involve oriented divisions and crowd movements of cells during morphogenetic events. Cellular behavior that can be recognized as individual cell migration is very limited and short-ranged and likely results from cellular mixing due to the proliferation activity of the tissue. These cellular mechanisms resemble the strategy behind limb bud morphogenesis, suggesting the possibility of common principles and deep homology between facial and limb outgrowth.


Asunto(s)
Diferenciación Celular , Células Clonales/citología , Cara/embriología , Morfogénesis , Cresta Neural/citología , Organogénesis , Animales , Movimiento Celular , Ectodermo/citología , Ectodermo/embriología , Expresión Génica , Genes Reporteros , Imagenología Tridimensional , Mesodermo/citología , Mesodermo/embriología , Ratones , Modelos Anatómicos , Fenotipo , Pez Cebra
17.
Nat Commun ; 7: 12186, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27435623

RESUMEN

Fast nerve conduction relies on successive myelin segments that electrically isolate axons. Segment geometry-diameter and length-is critical for the optimization of nerve conduction and the molecular mechanisms allowing this optimized geometry are partially known. We show here that peripheral myelin elongation is dynamically regulated by stimulation of YAP (Yes-associated protein) transcription cofactor activity during axonal elongation and limited by inhibition of YAP activity via the Hippo pathway. YAP promotes myelin and non-myelin genes transcription while the polarity protein Crb3, localized at the tips of the myelin sheath, activates the Hippo pathway to temper YAP activity, therefore allowing for optimal myelin growth. Dystrophic Dy(2j/2j) mice mimicking human peripheral neuropathy with reduced internodal lengths have decreased nuclear YAP which, when corrected, leads to longer internodes. These data show a novel mechanism controlling myelin growth and nerve conduction, and provide a molecular ground for disease with short myelin segments.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Axones/metabolismo , Proteínas de la Membrana/metabolismo , Vaina de Mielina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Axones/ultraestructura , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Células HEK293 , Vía de Señalización Hippo , Humanos , Glicoproteínas de Membrana , Ratones , Fenotipo , Fosforilación , Ratas , Células de Schwann/metabolismo , Transcripción Genética , Proteínas Señalizadoras YAP
18.
Nat Neurosci ; 19(8): 1050-1059, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27294512

RESUMEN

Schwann cell development and peripheral nerve myelination require the serial expression of transcriptional activators, such as Sox10, Oct6 (also called Scip or Pou3f1) and Krox20 (also called Egr2). Here we show that transcriptional repression, mediated by the zinc-finger protein Zeb2 (also known as Sip1), is essential for differentiation and myelination. Mice lacking Zeb2 in Schwann cells develop a severe peripheral neuropathy, caused by failure of axonal sorting and virtual absence of myelin membranes. Zeb2-deficient Schwann cells continuously express repressors of lineage progression. Moreover, genes for negative regulators of maturation such as Sox2 and Ednrb emerge as Zeb2 target genes, supporting its function as an 'inhibitor of inhibitors' in myelination control. When Zeb2 is deleted in adult mice, Schwann cells readily dedifferentiate following peripheral nerve injury and become repair cells. However, nerve regeneration and remyelination are both perturbed, demonstrating that Zeb2, although undetectable in adult Schwann cells, has a latent function throughout life.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Homeodominio/genética , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Proteínas Represoras/genética , Células de Schwann/metabolismo , Animales , Axones/metabolismo , Axones/ultraestructura , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Ratones Transgénicos , Nervios Periféricos/metabolismo , Células de Schwann/citología , Factores de Transcripción/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
19.
Nat Commun ; 6: 8584, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26466203

RESUMEN

MicroRNAs (miRNAs) are crucial regulators of myelination in the peripheral nervous system (PNS). However, the miRNAs species involved and the underlying mechanisms are largely unknown. We found that let-7 miRNAs are highly abundant during PNS myelination and that their levels are inversely correlated to the expression of lin28 homolog B (Lin28B), an antagonist of let-7 accumulation. Sustained expression of Lin28B and consequently reduced levels of let-7 miRNAs results in a failure of Schwann cell myelination in transgenic mouse models and in cell culture. Subsequent analyses revealed that let-7 miRNAs promote expression of the myelination-driving master transcription factor Krox20 (also known as Egr2) through suppression of myelination inhibitory Notch signalling. We conclude that the Lin28B/let-7 axis acts as a critical driver of PNS myelination, in particular by regulating myelination onset, identifying this pathway also as a potential therapeutic target in demyelinating diseases.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , MicroARNs/metabolismo , Vaina de Mielina/metabolismo , Sistema Nervioso Periférico/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al ARN , Ratas , Receptores Notch/metabolismo , Ribonucleasa III/metabolismo
20.
PLoS Biol ; 13(9): e1002258, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26406915

RESUMEN

The pathogenesis of peripheral neuropathies in adults is linked to maintenance mechanisms that are not well understood. Here, we elucidate a novel critical maintenance mechanism for Schwann cell (SC)-axon interaction. Using mouse genetics, ablation of the transcriptional regulators histone deacetylases 1 and 2 (HDAC1/2) in adult SCs severely affected paranodal and nodal integrity and led to demyelination/remyelination. Expression levels of the HDAC1/2 target gene myelin protein zero (P0) were reduced by half, accompanied by altered localization and stability of neurofascin (NFasc)155, NFasc186, and loss of Caspr and septate-like junctions. We identify P0 as a novel binding partner of NFasc155 and NFasc186, both in vivo and by in vitro adhesion assay. Furthermore, we demonstrate that HDAC1/2-dependent P0 expression is crucial for the maintenance of paranodal/nodal integrity and axonal function through interaction of P0 with neurofascins. In addition, we show that the latter mechanism is impaired by some P0 mutations that lead to late onset Charcot-Marie-Tooth disease.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Enfermedad de Charcot-Marie-Tooth/genética , Proteína P0 de la Mielina/genética , Vaina de Mielina/fisiología , Factores de Crecimiento Nervioso/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Enfermedad de Charcot-Marie-Tooth/enzimología , Técnicas de Inactivación de Genes , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...