Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38701782

RESUMEN

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Asunto(s)
Epigénesis Genética , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Vaina de Mielina/metabolismo , Humanos , Ratones , Remielinización/efectos de los fármacos , Oligodendroglía/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Endogámicos C57BL , Rejuvenecimiento , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Organoides/metabolismo , Organoides/efectos de los fármacos , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/genética , Diferenciación Celular/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Masculino , Regeneración/efectos de los fármacos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología
2.
J Neurosci ; 44(20)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38565291

RESUMEN

Microglia undergo two-stage activation in neurodegenerative diseases, known as disease-associated microglia (DAM). TREM2 mediates the DAM2 stage transition, but what regulates the first DAM1 stage transition is unknown. We report that glucose dyshomeostasis inhibits DAM1 activation and PKM2 plays a role. As in tumors, PKM2 was aberrantly elevated in both male and female human AD brains, but unlike in tumors, it is expressed as active tetramers, as well as among TREM2+ microglia surrounding plaques in 5XFAD male and female mice. snRNAseq analyses of microglia without Pkm2 in 5XFAD mice revealed significant increases in DAM1 markers in a distinct metabolic cluster, which is enriched in genes for glucose metabolism, DAM1, and AD risk. 5XFAD mice incidentally exhibited a significant reduction in amyloid pathology without microglial Pkm2 Surprisingly, microglia in 5XFAD without Pkm2 exhibited increases in glycolysis and spare respiratory capacity, which correlated with restoration of mitochondrial cristae alterations. In addition, in situ spatial metabolomics of plaque-bearing microglia revealed an increase in respiratory activity. These results together suggest that it is not only glycolytic but also respiratory inputs that are critical to the development of DAM signatures in 5XFAD mice.


Asunto(s)
Glucosa , Homeostasis , Ratones Transgénicos , Microglía , Animales , Microglía/metabolismo , Microglía/patología , Ratones , Homeostasis/fisiología , Glucosa/metabolismo , Masculino , Femenino , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glucólisis/fisiología , Proteínas de Unión a Hormona Tiroide
3.
J Neurosci ; 41(42): 8710-8724, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34507952

RESUMEN

We report that the neurotrophin receptor p75 contributes to sensory neuron survival through the regulation of cholesterol metabolism in Schwann cells. Selective deletion of p75 in mouse Schwann cells of either sex resulted in a 30% loss of dorsal root ganglia (DRG) neurons and diminished thermal sensitivity. P75 regulates Schwann cell cholesterol biosynthesis in response to BDNF, forming a co-receptor complex with ErbB2 and activating ErbB2-mediated stimulation of sterol regulatory element binding protein 2 (SREBP2), a master regulator of cholesterol synthesis. Schwann cells lacking p75 exhibited decreased activation of SREBP2 and a reduction in 7-dehydrocholesterol (7-DHC) reductase (DHCR7) expression, resulting in accumulation of the neurotoxic intermediate, 7-dehyrocholesterol in the sciatic nerve. Restoration of DHCR7 in p75 null Schwann cells in mice significantly attenuated DRG neuron loss. Together, these results reveal a mechanism by which the disruption of lipid metabolism in glial cells negatively influences sensory neuron survival, which has implications for a wide range of peripheral neuropathies.SIGNIFICANCE STATEMENT Although expressed in Schwann cells, the role of p75 in myelination has remained unresolved in part because of its dual expression in sensory neurons that Schwann cells myelinate. When p75 was deleted selectively among Schwann cells, myelination was minimally affected, while sensory neuron survival was reduced by 30%. The phenotype is mainly due to dysregulation of cholesterol biosynthesis in p75-deficient Schwann cells, leading to an accumulation of neurotoxic cholesterol precursor, 7-dehydrocholesterol (7-DHC). Mechanism-wise, we discovered that in response to BDNF, p75 recruits and activates ErbB2 independently of ErbB3, thereby stimulating the master regulator, sterol regulatory element binding protein 2 (SREBP2). These results together highlight a novel role of p75 in Schwann cells in regulating DRG neuron survival by orchestrating proper cholesterol metabolism.


Asunto(s)
Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Células de Schwann/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Células de Schwann/ultraestructura , Células Receptoras Sensoriales/ultraestructura
4.
Dev Biol ; 462(1): 36-49, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32113830

RESUMEN

Embryonic morphogenesis relies on the intrinsic ability of cells, often through remodeling the cytoskeleton, to shape epithelial tissues during development. Epithelial invagination is an example of morphogenesis that depends on this remodeling but the cellular mechanisms driving arrangement of cytoskeletal elements needed for tissue deformation remain incompletely characterized. To elucidate these mechanisms, live fluorescent microscopy and immunohistochemistry on fixed specimens were performed on chick and mouse lens placodes. This analysis revealed the formation of peripherally localized, circumferentially orientated and aligned junctions enriched in F-actin and MyoIIB. Once formed, the aligned junctions contract in a Rho-kinase and non-muscle myosin dependent manner. Further molecular characterization of these junctions revealed a Rho-kinase dependent accumulation of Arhgef11, a RhoA-specific guanine exchange factor known to regulate the formation of actomyosin cables and junctional contraction. In contrast, the localization of the Par-complex protein Par3, was reduced in these circumferentially orientated junctions. In an effort to determine if Par3 plays a negative role in MyoIIB accumulation, Par3-deficient mouse embryos were analyzed which not only revealed an increase in bicellular junctional accumulation of MyoIIB, but also a reduction of Arhgef11. Together, these results highlight the importance of the formation of the multicellular actomyosin cables that appear essential to the initiation of epithelial invagination and implicate the potential role of Arhgef11 and Par3 in their contraction and formation.


Asunto(s)
Actomiosina/metabolismo , Cristalino/embriología , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Uniones Adherentes/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Embrión de Pollo , Citoesqueleto/metabolismo , Desarrollo Embrionario , Células Epiteliales/metabolismo , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Ratones Noqueados , Morfogénesis , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Quinasas Asociadas a rho/metabolismo
5.
Anal Chem ; 91(3): 2304-2311, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30608652

RESUMEN

Sensitivity and resolution are key considerations for NMR applications in general and for metabolomics in particular, where complex mixtures containing hundreds of metabolites over a large range of concentrations are commonly encountered. There is a strong demand for advanced methods that can provide maximal information in the shortest possible time frame. Here, we present the optimization and application of the recently introduced 2D real-time BIRD 1H-13C HSQC experiment for NMR-based metabolomics of aqueous samples at 13C natural abundance. For mouse urine samples, it is demonstrated how this real-time pure shift sensitivity-improved heteronuclear single quantum correlation method provides broadband homonuclear decoupling along the proton detection dimension and thereby significantly improves spectral resolution in regions that are affected by spectral overlap. Moreover, the collapse of the scalar multiplet structure of cross-peaks leads to a sensitivity gain of about 40-50% over a traditional 2D HSQC-SI experiment. The experiment works well over a range of magnetic field strengths and is particularly useful when resonance overlap in crowded regions of the HSQC spectra hampers accurate metabolite identification and quantitation.


Asunto(s)
Metaboloma , Metabolómica/métodos , Resonancia Magnética Nuclear Biomolecular/métodos , Orina/química , Animales , Ratones
6.
J Biol Chem ; 289(45): 31458-72, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25253694

RESUMEN

p75 is expressed among Purkinje cells in the adult cerebellum, but its function has remained obscure. Here we report that p75 is involved in maintaining the frequency and regularity of spontaneous firing of Purkinje cells. The overall spontaneous firing activity of Purkinje cells was increased in p75(-/-) mice during the phasic firing period due to a longer firing period and accompanying reduction in silence period than in the wild type. We attribute these effects to a reduction in small conductance Ca(2+)-activated potassium (SK) channel activity in Purkinje cells from p75(-/-) mice compared with the wild type littermates. The mechanism by which p75 regulates SK channel activity appears to involve its ability to activate Rac1. In organotypic cultures of cerebellar slices, brain-derived neurotrophic factor increased RacGTP levels by activating p75 but not TrkB. These results correlate with a reduction in RacGTP levels in synaptosome fractions from the p75(-/-) cerebellum, but not in that from the cortex of the same animals, compared with wild type littermates. More importantly, we demonstrate that Rac1 modulates SK channel activity and firing patterns of Purkinje cells. Along with the finding that spine density was reduced in p75(-/-) cerebellum, these data suggest that p75 plays a role in maintaining normalcy of Purkinje cell firing in the cerebellum in part by activating Rac1 in synaptic compartments and modulating SK channels.


Asunto(s)
Cerebelo/metabolismo , Neuropéptidos/metabolismo , Células de Purkinje/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Electrofisiología , Aparato de Golgi/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Sinaptosomas/metabolismo , Tetraetilamonio/química , Proteínas de Unión al GTP rac/metabolismo
7.
EMBO J ; 30(22): 4665-77, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21964071

RESUMEN

Subcellular localization of mRNAs is regulated by RNA-protein interactions. Here, we show that introduction of a reporter mRNA with the 3'UTR of ß-actin mRNA competes with endogenous mRNAs for binding to ZBP1 in adult sensory neurons. ZBP1 is needed for axonal localization of ß-actin mRNA, and introducing GFP with the 3'UTR of ß-actin mRNA depletes axons of endogenous ß-actin and GAP-43 mRNAs and attenuates both in vitro and in vivo regrowth of severed axons. Consistent with limited levels of ZBP1 protein in adult neurons, mice heterozygous for the ZBP1 gene are haploinsufficient for axonal transport of ß-actin and GAP-43 mRNAs and for regeneration of peripheral nerve. Exogenous ZBP1 can rescue the RNA transport deficits, but the axonal growth deficit is only rescued if the transported mRNAs are locally translated. These data support a direct role for ZBP1 in transport and translation of mRNA cargos in axonal regeneration in vitro and in vivo.


Asunto(s)
Actinas/genética , Axones/fisiología , Glicoproteínas/metabolismo , Regeneración Nerviosa/fisiología , ARN Mensajero/metabolismo , Regiones no Traducidas 3'/genética , Actinas/metabolismo , Animales , Transporte Axonal/genética , Proliferación Celular , Células Cultivadas , Proteína GAP-43/deficiencia , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/genética , Conos de Crecimiento/fisiología , Ratones , Ratones Endogámicos C57BL , Transporte de ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/metabolismo
8.
J Neurosci ; 33(6): 2388-97, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23392668

RESUMEN

In the developing peripheral nervous system, axon-derived signals stimulate Schwann cells to undergo a global genetic reprogramming involving the cessation of cellular division and the upregulation of myelin genes. How such a comprehensive change in gene transcription is regulated is poorly understood. Here we report that BRG1/SMARCA4, the central helicase of the mammalian SWI/SNF-related chromatin remodeling complex, is required for Schwann cells to differentiate and form myelin, both in vitro and in vivo, in the mouse. BRG1 was highly activated in Schwann cells at early stages of myelination, and loss of the enzyme inhibited their differentiation and completely prevented myelin formation. Furthermore, we identify NF-κB as a key transcription factor that associates with the BRG1 complex in response to neuregulin 1 type III. During myelination, BRG1 was activated through the formation of a complex with NF-κB, and both proteins bound to the promoter region of Sox10, an inducer of myelination. These findings delineate a novel mechanism whereby axonal signals promote myelination through the remodeling of chromatin structure.


Asunto(s)
Diferenciación Celular/fisiología , Cromatina/metabolismo , ADN Helicasas/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Células de Schwann/fisiología , Factores de Transcripción/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Cromatina/fisiología , Técnicas de Cocultivo , ADN Helicasas/fisiología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/fisiología , Proteínas Nucleares/fisiología , Ratas , Células de Schwann/citología , Factores de Transcripción/fisiología
9.
J Neurosci ; 33(2): 397-410, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23303920

RESUMEN

The lack of effective therapies for spinal cord injury points to the need for identifying novel targets for therapeutic intervention. Here we report that a small molecule, LM11A-31, developed to block proNGF-p75 interaction and p75-mediated cell death crosses the blood-brain barrier efficiently when delivered orally. Administered starting 4 h postinjury, LM11A-31 promotes functional recovery without causing any toxicity or increased pain in a mouse model of spinal contusion injury. In both weight-bearing open-field tests and nonweight-bearing swim tests, LM11A-31 was effective in improving motor function and coordination. Such functional improvement correlated with a >50% increase in the number of surviving oligodendrocytes and myelinated axons. We also demonstrate that LM11A-31 indeed inhibits proNGF-p75 interaction in vivo, thereby curtailing the JNK3-mediated apoptotic cascade. These results thus highlight p75 as a novel therapeutic target for an orally delivered treatment for spinal cord injury.


Asunto(s)
Isoleucina/análogos & derivados , Morfolinas/uso terapéutico , Vaina de Mielina/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Receptor de Factor de Crecimiento Nervioso/efectos de los fármacos , Receptor de Factor de Crecimiento Nervioso/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Western Blotting , ADN/genética , Relación Dosis-Respuesta a Droga , Miembro Anterior/fisiología , Miembro Posterior/fisiología , Hiperalgesia/tratamiento farmacológico , Inmunohistoquímica , Isoleucina/uso terapéutico , Locomoción/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Reacción en Cadena de la Polimerasa , Traumatismos de la Médula Espinal/patología , Natación/fisiología
10.
J Biol Chem ; 287(2): 1600-8, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22128191

RESUMEN

Brain-derived neurotrophic factor (BDNF) was shown to play a role in Schwann cell myelination by recruiting Par3 to the axon-glial interface, but the underlying mechanism has remained unclear. Here we report that Par3 regulates Rac1 activation by BDNF but not by NRG1-Type III in Schwann cells, although both ligands activate Rac1 in vivo. During development, active Rac1 signaling is localized to the axon-glial interface in Schwann cells by a Par3-dependent polarization mechanism. Knockdown of p75 and Par3 individually inhibits Rac1 activation, whereas constitutive activation of Rac1 disturbs the polarized activation of Rac1 in vivo. Polarized Rac1 activation is necessary for myelination as Par3 knockdown attenuates myelination in mouse sciatic nerves as well as in zebrafish. Specifically, Par3 knockdown in zebrafish disrupts proper alignment between the axon and Schwann cells without perturbing Schwann cell migration, suggesting that localized Rac1 activation at the axon-glial interface helps identify the initial wrapping sites. We therefore conclude that polarization of Rac1 activation is critical for myelination.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Moléculas de Adhesión Celular/metabolismo , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Células de Schwann/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas Portadoras/genética , Moléculas de Adhesión Celular/genética , Proteínas de Ciclo Celular , Movimiento Celular/fisiología , Células Cultivadas , Ratones , Proteínas del Tejido Nervioso/genética , Neuroglía/citología , Neuroglía/metabolismo , Neuropéptidos/genética , Ratas , Células de Schwann/citología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética
11.
Cerebellum ; 12(3): 300-3, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23307658

RESUMEN

The p75 neurotrophin receptor is highly expressed in the developing nervous system and is required for neuronal survival, growth, and synaptic transmission. In young mice, p75 is present in both granular cells and Purkinje cells of the cerebellum. Although p75 has been implicated in modulation of neuronal excitability in several neuronal types, whether and how it affects the excitability of cerebellar Purkinje neurons remained unclear. Using extracellular recordings of spontaneous firing of Purkinje neurons in cerebellar slices prepared from wild type and p75 knockout mice, we measured intrinsic firing properties in the presence of fast synaptic blockers of more than 200 Purkinje cells, each for a period of 5 min, for each genotype. We detected a significant increase in the mean firing frequency in p75(-/-) neurons comparing to the wild type littermates. Upon separating tonically firing from phasically firing cells, i.e., cells with firing pauses of longer than 300 ms, we observed that the change mainly arose from phasic firing cells and can be explained by an increase in the firing/silence ratio and a decrease in the number of long pauses during the 5-min recording period. We conclude that p75 plays an important role in regulating the firing-to-silence transition during the phasic firing period of the spontaneous firing of Purkinje cells. Thus, p75 exerts a modulatory function on Purkinje cell firing patterns, through which it may act as a key player in motor coordination and other cerebellum-regulated activities since Purkinje cells represent the sole neuronal output of the cerebellar cortex.


Asunto(s)
Potenciales de Acción/genética , Cerebelo/citología , Células de Purkinje/fisiología , Receptores de Factor de Crecimiento Nervioso/deficiencia , Animales , Animales Recién Nacidos , Técnicas In Vitro , Ratones , Ratones Noqueados
12.
J Neurosci ; 31(41): 14481-7, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21994364

RESUMEN

Axonal mRNA transport is robust in cultured neurons but there has been limited evidence for this in vivo. We have used a genetic approach to test for in vivo axonal transport of reporter mRNAs. We show that ß-actin's 3'-UTR can drive axonal localization of GFP mRNA in mature DRG neurons, but mice with γ-actin's 3'-UTR show no axonal GFP mRNA. Peripheral axotomy triggers transport of the ß-actin 3'-UTR containing transgene mRNA into axons. This GFP-3'-ß-actin mRNA accumulates in injured PNS axons before activation of the transgene promoter peaks in the DRG. Spinal cord injury also increases axonal GFP signals in mice carrying this transgene without any increase in transgene expression in the DRGs. These data show for the first time that the ß-actin 3'-UTR is sufficient for axonal localization in both PNS and CNS neurons in vivo.


Asunto(s)
Axones/metabolismo , Ganglios Espinales/citología , Neuronas/citología , ARN Mensajero/metabolismo , Médula Espinal/citología , Regiones no Traducidas 3'/genética , Actinas/genética , Actinas/metabolismo , Análisis de Varianza , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas , Dendritas/metabolismo , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Microscopía Confocal/métodos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/genética , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , Células de Schwann/metabolismo , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
13.
J Biol Chem ; 285(26): 20358-68, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20421303

RESUMEN

During the development of the sympathetic nervous system, the p75 neurotrophin receptor (p75NTR) has a dual function: promoting survival together with TrkA in response to NGF, but inducing cell death upon binding pro or mature brain-derived neurotrophic factor (BDNF). Apoptotic signaling through p75NTR requires activation of the stress kinase, JNK. However, the receptor also undergoes regulated proteolysis, first by a metalloprotease, and then by gamma-secretase, in response to pro-apoptotic ligands and this is necessary for receptor mediated neuronal death (Kenchappa, R. S., Zampieri, N., Chao, M. V., Barker, P. A., Teng, H. K., Hempstead, B. L., and Carter, B. D. (2006) Neuron 50, 219-232). Hence, the relationship between JNK activation and receptor proteolysis remains to be defined. Here, we report that JNK3 activation is necessary for p75NTR cleavage; however, following release of the intracellular domain, there is a secondary activation of JNK3 that is cleavage dependent. Receptor proteolysis and apoptosis were prevented in sympathetic neurons from jnk3(-/-) mice, while activation of JNK by ectopic expression of MEKK1 induced p75NTR cleavage and cell death. Proteolysis of the receptor was not detected until 6 h after BDNF treatment, suggesting that JNK3 promotes cleavage through a transcriptional mechanism. In support of this hypothesis, BDNF up-regulated tumor necrosis factor-alpha-converting enzyme (TACE)/ADAM17 mRNA and protein in wild-type, but not jnk3(-/-) sympathetic neurons. Down-regulation of TACE by RNA interference blocked BDNF-induced p75NTR cleavage and apoptosis, indicating that this metalloprotease is responsible for the initial processing of the receptor. Together, these results demonstrate that p75NTR-mediated activation of JNK3 is required for up-regulation of TACE, which promotes receptor proteolysis, leading to prolonged activation of JNK3 and subsequent apoptosis in sympathetic neurons.


Asunto(s)
Proteínas ADAM/metabolismo , Apoptosis , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Animales , Antracenos/farmacología , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/farmacología , Línea Celular , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Humanos , Cinética , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 10 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 10 Activada por Mitógenos/genética , Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores de Factor de Crecimiento Nervioso/genética , Ganglio Cervical Superior/citología , Regulación hacia Arriba
14.
ACS Med Chem Lett ; 12(10): 1546-1552, 2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34676036

RESUMEN

An indazole/aza-indazole scaffold was developed as a novel chemotype for JNK3 inhibition. Extensive structure activity relationship (SAR) studies utilizing various in vitro and in vivo assays led to potent and highly selective JNK3 inhibitors with good oral bioavailability and high brain penetration. One lead compound, 29, was a potent and selective JNK3 inhibitor (IC50 = 0.005 µM) that had significant inhibition (>80% at 1 µM) to only JNK3 and JNK2 in a panel profiling of 374 wild-type kinases, had high potency in functional cell-based assays, had high stability in the human liver microsome (t 1/2 = 92 min), and was orally bioavailable and brain penetrant (brain/plasma ratio: 56%). The cocrystal structure of 29 in human JNK3 at a 2.1 Å resolution showed that indazole or aza-indazole-based JNK3 inhibitors demonstrated a type I kinase inhibition/binding.

15.
ACS Med Chem Lett ; 12(1): 24-29, 2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33488960

RESUMEN

Potent JNK3 isoform selective inhibitors were developed from a thiophenyl-pyrazolourea scaffold. Through structure activity relationship (SAR) studies utilizing enzymatic and cell-based assays, and in vitro and in vivo drug metabolism and pharmacokinetic (DMPK) studies, potent and highly selective JNK3 inhibitors with oral bioavailability and brain penetrant capability were developed. Inhibitor 17 was a potent and isoform selective JNK3 inhibitor (IC50 = 35 nM), had significant inhibition to only JNK3 in a panel profiling of 374 wild-type kinases, had high potency in functional cell-based assays, had high stability in human liver microsome (t 1/2 = 66 min) and a clean CYP-450 inhibition profile, and was orally bioavailable and brain penetrant. Moreover, cocrystal structures of compounds 17 and 27 in human JNK3 were solved at 1.84 Å, which showed that these JNK3 isoform selective inhibitors bound to the ATP pocket, had interactions in both hydrophobic pocket-I and hydrophobic pocket-II.

16.
Circ Res ; 103(2): e15-26, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18566344

RESUMEN

Diabetes impairs endothelial function and reparative neovascularization. The p75 receptor of neurotrophins (p75(NTR)), which is scarcely present in healthy endothelial cells (ECs), becomes strongly expressed by capillary ECs after induction of peripheral ischemia in type-1 diabetic mice. Here, we show that gene transfer-induced p75(NTR) expression impairs the survival, proliferation, migration, and adhesion capacities of cultured ECs and endothelial progenitor cells (EPCs) and inhibits angiogenesis in vitro. Moreover, intramuscular p75(NTR) gene delivery impairs neovascularization and blood flow recovery in a mouse model of limb ischemia. These disturbed functions are associated with suppression of signaling mechanisms implicated in EC survival and angiogenesis. In fact, p75(NTR) depresses the VEGF-A/Akt/eNOS/NO pathway and additionally reduces the mRNA levels of ITGB1 [beta (1) integrin], BIRC5 (survivin), PTTG1 (securin) and VEZF1. Diabetic mice, which typically show impaired postischemic muscular neovascularization and blood perfusion recovery, have these defects corrected by intramuscular gene transfer of a dominant negative mutant form of p75(NTR). Collectively, our data newly demonstrate the antiangiogenic action of p75(NTR) and open new avenues for the therapeutic use of p75(NTR) inhibition to combat diabetes-induced microvascular liabilities.


Asunto(s)
Apoptosis/fisiología , Angiopatías Diabéticas/metabolismo , Endotelio Vascular/fisiología , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Humanos , Isquemia/etiología , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Factor de Crecimiento Nervioso/genética , Transducción de Señal/fisiología , Estreptozocina , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Elife ; 92020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31995032

RESUMEN

Networks of neurons control feeding and activity patterns by integrating internal metabolic signals of energy balance with external environmental cues such as time-of-day. Proper circadian alignment of feeding behavior is necessary to prevent metabolic disease, and thus it is imperative that molecular players that maintain neuronal coordination of energy homeostasis are identified. Here, we demonstrate that mice lacking the p75 neurotrophin receptor, p75NTR, decrease their feeding and food anticipatory behavior (FAA) in response to daytime, but not nighttime, restricted feeding. These effects lead to increased weight loss, but do not require p75NTR during development. Instead, p75NTR is required for fasting-induced activation of neurons within the arcuate hypothalamus. Indeed, p75NTR specifically in AgRP neurons is required for FAA in response to daytime restricted feeding. These findings establish p75NTR as a novel regulator gating behavioral response to food scarcity and time-of-day dependence of circadian food anticipation.


In many animals, specific types of neurons, such as the hypothalamic hunger neurons, are tasked with regulating food consumption, integrating internal signals of hunger. In general, individuals eat if food becomes available when they are hungry; if food is absent, they will start moving to find new resources. Finally, if food always comes at the same time, animals will increase their activity just before it is delivered. Neurotrophins are a family of proteins that have many essential roles in the brain. In recent years, they have been shown to interact with the circadian clock, the built-in mechanism that helps animals stay synchronized with the cycle of day and night. A protein known as p75NTR is present in nerve cells, including hypothalamic hunger neurons: there, it helps to relay messages from a neurotrophin which, amongst other roles, controls food intake. However, it was unclear whether p75NTR played a role in regulating feeding behaviors, especially in a circadian manner. To investigate this question, Podyma et al. genetically engineered a group of mice lacking p75NTR, and a group missing the protein only in their hypothalamic hunger neurons. Both types of mutants had abnormal control of their feeding behavior: compared to normal mice, they fed less (and lost more weight) after they had been deprived of food overnight, or when they faced food shortage over multiple days. In addition, the mutants failed to move more before being fed. However, these feeding patterns were only affected during daytime, while they were preserved at night. These results reveal a new role for p75NTR in hypothalamic hunger neurons. Dissecting the biological processes that control food intake is key since obesity levels are increasing around the world. In particular, the relationship between food intake and the circadian clock is an important avenue of research as time-restricted diets (where food intake is only allowed during specific periods of the day) are growing in popularity.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Conducta Alimentaria , Homeostasis , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Análisis Químico de la Sangre , Ritmo Circadiano , Mutación de Línea Germinal , Ratones , Ratones Noqueados , Receptores de Factor de Crecimiento Nervioso/genética
18.
Curr Biol ; 16(24): 2446-52, 2006 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-17174920

RESUMEN

A central pathogenic feature of neurodegenerative diseases and neurotrauma is the death of neurons. A mechanistic understanding of the factors and conditions that induce the dysfunction and death of neurons is essential for devising effective treatment strategies against neuronal loss after trauma or during aging. Because Apolipoprotein E (ApoE) is a major risk factor for several neurodegenerative diseases, including Alzheimer's disease , a direct or indirect role of ApoE receptors in the disease process is likely. Here we have used gene targeting in mice to investigate possible roles of ApoE receptors in the regulation of neuronal survival. We demonstrate that a differentially spliced isoform of an ApoE receptor, ApoE receptor 2 (Apoer2), is essential for protection against neuronal cell loss during normal aging. Furthermore, the same splice form selectively promotes neuronal cell death after injury through mechanisms that may involve serine/threonine kinases of the Jun N-terminal kinase (JNK) family. These findings raise the possibility that ApoE and its receptors cooperatively regulate common mechanisms that are essential to neuronal survival in the adult brain.


Asunto(s)
Encéfalo/fisiología , Neuronas/citología , Receptores de Lipoproteína/fisiología , Envejecimiento , Empalme Alternativo , Animales , Apolipoproteínas E/fisiología , Encéfalo/citología , Muerte Celular , Supervivencia Celular , Exones , Proteínas Relacionadas con Receptor de LDL , Ratones , Ratones Noqueados , Proteína Quinasa 10 Activada por Mitógenos/genética , Neuronas/fisiología , Estructura Terciaria de Proteína , Receptores de Lipoproteína/genética
19.
Neuron ; 36(3): 375-86, 2002 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-12408842

RESUMEN

The neurotrophin receptor p75 is induced by various injuries to the nervous system, but its role after injury has remained unclear. Here, we report that p75 is required for the death of oligodendrocytes following spinal cord injury, and its action is mediated mainly by proNGF. Oligodendrocytes undergoing apoptosis expressed p75, and the absence of p75 resulted in a decrease in the number of apoptotic oligodendrocytes and increased survival of oligodendrocytes. ProNGF is likely responsible for activating p75 in vivo, since the proNGF from the injured spinal cord induced apoptosis among p75(+/+), but not among p75(-/-), oligodendrocytes in culture, and its action was blocked by proNGF-specific antibody. Together, these data suggest that the role of proNGF is to eliminate damaged cells by activating the apoptotic machinery of p75 after injury.


Asunto(s)
Apoptosis/genética , Péptidos y Proteínas de Señalización Intracelular , Factor de Crecimiento Nervioso/metabolismo , Oligodendroglía/metabolismo , Precursores de Proteínas/metabolismo , Receptor de Factor de Crecimiento Nervioso/deficiencia , Traumatismos de la Médula Espinal/metabolismo , Animales , Especificidad de Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Proteínas Relacionadas con la Autofagia , Caspasa 3 , Caspasas/metabolismo , Supervivencia Celular/fisiología , Femenino , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Factor de Crecimiento Nervioso/inmunología , Factor de Crecimiento Nervioso/farmacología , Precursores de Proteínas/inmunología , Precursores de Proteínas/farmacología , Proteínas/metabolismo , Tiempo de Reacción/fisiología , Receptor de Factor de Crecimiento Nervioso/efectos de los fármacos , Receptor de Factor de Crecimiento Nervioso/genética , Proteínas Recombinantes de Fusión , Traumatismos de la Médula Espinal/fisiopatología
20.
J Neurosci ; 27(31): 8395-404, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17670986

RESUMEN

Although oligodendrocytes undergo apoptosis after spinal cord injury, molecular mechanisms responsible for their death have been unknown. We report that oligodendrocyte apoptosis is regulated oppositely by c-Jun N-terminal kinase 3 (JNK3) and protein interacting with the mitotic kinase, never in mitosis A I (Pin1), the actions of which converge on myeloid cell leukemia sequence-1 (Mcl-1). Activated after injury, JNK3 induces cytochrome c release by facilitating the degradation of Mcl-1, the stability of which is maintained in part by Pin1. Pin1 binds Mcl-1 at its constitutively phosphorylated site, Thr163Pro, and stabilizes it by inhibiting ubiquitination. After injury JNK3 phosphorylates Mcl-1 at Ser121Pro, facilitating the dissociation of Pin1 from Mcl-1. JNK3 thus induces Mcl-1 degradation by counteracting the protective binding of Pin1. These results are confirmed by the opposing phenotypes observed between JNK3-/- and Pin1-/- mice: oligodendrocyte apoptosis and cytochrome c release are reduced in JNK3-/- but elevated in Pin1-/- mice. This report thus unveils a mechanism by which cytochrome c release is under the opposite control of JNK3 and Pin1, regulators for which the activities are intricately coupled.


Asunto(s)
Apoptosis/fisiología , Proteína Quinasa 10 Activada por Mitógenos/fisiología , Oligodendroglía/enzimología , Isomerasa de Peptidilprolil/fisiología , Traumatismos de la Médula Espinal/enzimología , Animales , Apoptosis/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 10 Activada por Mitógenos/deficiencia , Proteína Quinasa 10 Activada por Mitógenos/genética , Peptidilprolil Isomerasa de Interacción con NIMA , Oligodendroglía/citología , Oligodendroglía/patología , Isomerasa de Peptidilprolil/deficiencia , Isomerasa de Peptidilprolil/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Ubiquitina/antagonistas & inhibidores , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA