Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(24): 12035-12044, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31138690

RESUMEN

PSD-95 is a scaffolding protein that regulates the synaptic localization of many receptors, channels, and signaling proteins. The NLGN gene family encodes single-pass transmembrane postsynaptic cell adhesion molecules that are important for synapse assembly and function. At excitatory synapses, NLGN1 mediates transsynaptic binding with neurexin, a presynaptic cell adhesion molecule, and also binds to PSD-95, although the relevance of the PSD-95 interaction is not clear. We now show that disruption of the NLGN1 and PSD-95 interaction decreases surface expression of NLGN1 in cultured neurons. Furthermore, PKA phosphorylates NLGN1 on S839, near the PDZ ligand, and dynamically regulates PSD-95 binding. A phosphomimetic mutation of NLGN1 S839 significantly reduced PSD-95 binding. Impaired NLGN1/PSD-95 binding diminished synaptic NLGN1 expression and NLGN1-mediated synaptic enhancement. Our results establish a phosphorylation-dependent molecular mechanism that regulates NLGN1 and PSD-95 binding and provides insights into excitatory synaptic development and function.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Animales , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Fosforilación/fisiología , Unión Proteica/fisiología , Sinapsis/metabolismo
2.
J Biol Chem ; 294(30): 11498-11512, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31177092

RESUMEN

Neurolastin is a dynamin family GTPase that also contains a RING domain and exhibits both GTPase and E3 ligase activities. It is specifically expressed in the brain and is important for synaptic transmission, as neurolastin knockout animals have fewer dendritic spines and exhibit a reduction in functional synapses. Our initial study of neurolastin revealed that it is membrane-associated and partially co-localizes with endosomes. Using various biochemical and cell-culture approaches, we now show that neurolastin also localizes to mitochondria in HeLa cells, cultured neurons, and brain tissue. We found that the mitochondrial localization of neurolastin depends upon an N-terminal mitochondrial targeting sequence and that neurolastin is imported into the mitochondrial intermembrane space. Although neurolastin was only partially mitochondrially localized at steady state, it displayed increased translocation to mitochondria in response to neuronal stress and mitochondrial fragmentation. Interestingly, inactivation or deletion of neurolastin's RING domain also increased its mitochondrial localization. Using EM, we observed that neurolastin knockout animals have smaller but more numerous mitochondria in cerebellar Purkinje neurons, indicating that neurolastin regulates mitochondrial morphology. We conclude that the brain-specific dynamin GTPase neurolastin exhibits stress-responsive localization to mitochondria and is required for proper mitochondrial morphology.


Asunto(s)
Dinaminas/metabolismo , Mitocondrias/metabolismo , Células de Purkinje/metabolismo , Animales , Células Cultivadas , Dinaminas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/enzimología , Mutación , Transporte de Proteínas
3.
J Neurosci ; 37(15): 4093-4102, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28283559

RESUMEN

NMDA receptors (NMDARs) are ionotropic glutamate receptors that are crucial for neuronal development and higher cognitive processes. NMDAR dysfunction is involved in a variety of neurological and psychiatric diseases; however, the mechanistic link between the human pathology and NMDAR dysfunction is poorly understood. Rare missense variants within NMDAR subunits have been identified in numerous patients with mental or neurological disorders. We specifically focused on the GluN2B NMDAR subunit, which is highly expressed in the hippocampus and cortex throughout development. We analyzed several variants located in the GluN2B C terminus and found that three variants in patients with autism (S1415L) or schizophrenia (L1424F and S1452F) (S1413L, L1422F, and S1450F in rodents, respectively) displayed impaired binding to membrane-associated guanylate kinase (MAGUK) proteins. In addition, we observed a deficit in surface expression for GluN2B S1413L. Furthermore, there were fewer dendritic spines in GluN2B S1413L-expressing neurons. Importantly, synaptic NMDAR currents in neurons transfected with GluN2B S1413L in GluN2A/B-deficient mouse brain slices revealed only partial rescue of synaptic current amplitude. Functional properties of GluN2B S1413L in recombinant systems revealed no change in receptor properties, consistent with synaptic defects being the result of reduced trafficking and targeting of GluN2B S1413L to the synapse. Therefore, we find that GluN2B S1413L displays deficits in NMDAR trafficking, synaptic currents, and spine density, raising the possibility that this mutation may contribute to the phenotype in this autism patient. More broadly, our research demonstrates that the targeted study of certain residues in NMDARs based on rare variants identified in patients is a powerful approach to studying receptor function.SIGNIFICANCE STATEMENT We have used a "bedside-to-bench" approach to investigate the functional regulation of NMDA receptors (NMDARs). Using information from deep sequencing of patients with neurological or psychiatric disorders, we investigated missense variants identified in the intracellular C-terminal domain of the GluN2B NMDAR subunit. We found several variants that displayed altered properties. In particular, one variant identified in a patient with autism, human GluN2B S1415L, displayed reduced surface expression and binding to PSD-95. Furthermore expression of GluN2B S1415L (S1413L in mouse) showed a deficit in rescue of synaptic NMDAR currents and fewer dendritic spines, consistent with other reports of spine abnormalities being associated with autism. More broadly, we demonstrate that using patient data is an effective approach to probing the structure/function relationship of NMDARs.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Variación Genética/genética , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/genética , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo
4.
Proc Natl Acad Sci U S A ; 108(37): 15219-24, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21896768

RESUMEN

Glutamate is the major excitatory neurotransmitter in the mammalian CNS and mediates fast synaptic transmission upon activation of glutamate-gated ion channels. In addition, glutamate modulates a variety of other synaptic responses and intracellular signaling by activating metabotropic glutamate receptors (mGluRs), which are G protein-coupled receptors. The mGluRs are also expressed in nonneuronal tissues and are implicated in a variety of normal biological functions as well as diseases. To study mGluR-activated calcium signaling in neurons, we generated mGluR5 transgenic animals using a Thy1 promoter to drive expression in the forebrain, and one founder unexpectedly developed melanoma. To directly investigate the role of mGluR5 in melanoma formation, we generated mGluR5 transgenic lines under a melanocyte-specific promoter, tyrosinase-related protein 1. A majority of the founders showed a severe phenotype with early onset. Hyperpigmentation of the pinnae and tail could be detected as early as 3-5 d after birth for most of the mGluR5 transgene-positive mice. There was 100% penetrance in the progeny from the tyrosinase-related protein 1-mGluR5 lines generated from founders that developed melanoma. Expression of mGluR5 was detected in melanoma samples by RT-PCR, immunoblotting, and immunohistochemistry. We evaluated the expression of several cancer-related proteins in tumor samples and observed a dramatic increase in the phosphorylation of ERK, implicating ERK as a downstream effector of mGluR5 signaling in tumors. Our findings show that mGluR5-mediated glutamatergic signaling can trigger melanoma in vivo. The aggressive growth and severe phenotype make these mouse lines unique and a potentially powerful tool for therapeutic studies.


Asunto(s)
Melanoma/metabolismo , Melanoma/patología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Receptores de Glutamato Metabotrópico/metabolismo , Envejecimiento/patología , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Melanoma/enzimología , Ratones , Ratones Transgénicos , Neoplasias de los Músculos/metabolismo , Neoplasias de los Músculos/patología , Invasividad Neoplásica , Pigmentación , Receptor del Glutamato Metabotropico 5 , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología
5.
J Biol Chem ; 285(27): 20975-81, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20427279

RESUMEN

N-Methyl-d-aspartate (NMDA) receptors are expressed at excitatory synapses throughout the brain and are essential for neuronal development and synaptic plasticity. Functional NMDA receptors are tetramers, typically composed of NR1 and NR2 subunits (NR2A-D). NR2A and NR2B are expressed in the forebrain and are thought to assemble as diheteromers (NR1/NR2A, NR1/NR2B) and triheteromers (NR1/NR2A/NR2B). NR2A and NR2B contain cytosolic domains that regulate distinct postendocytic sorting events, with NR2A sorting predominantly into the degradation pathway, and NR2B preferentially trafficking through the recycling pathway. However, the interplay between these two subunits remains an open question. We have now developed a novel approach based on the dimeric feature of the alpha- and beta-chains of the human major histocompatibility complex class II molecule. We created chimeras of alpha- and beta-chains with the NR2A and NR2B C termini and evaluated endocytosis of dimers. Like chimeric proteins containing only a single NR2A or NR2B C-terminal domain, major histocompatibility complex class II-NR2A homodimers sort predominantly to late endosomes, whereas NR2B homodimers traffic to recycling endosomes. Interestingly, NR2A/NR2B heterodimers traffic preferentially through the recycling pathway, and NR2B is dominant in regulating dimer trafficking in both heterologous cells and neurons. In addition, the recycling of NR2B-containing NMDARs in wild-type neurons is not significantly different from NR2A(-/-) neurons. These data support a dominant role for NR2B in regulating the trafficking of triheteromeric NMDARs in vivo. Furthermore, our molecular approach allows for the direct and selective evaluation of dimeric assemblies and can be used to define dominant trafficking domains in other multisubunit protein complexes.


Asunto(s)
Neuronas/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Anticuerpos Monoclonales , Células Cultivadas , ADN Complementario/genética , Dimerización , Endosomas/fisiología , Regulación de la Expresión Génica , Genes Reporteros , Antígenos HLA-DR/inmunología , Células HeLa/fisiología , Humanos , Ratones , Neuronas/citología , Prosencéfalo/fisiología , Subunidades de Proteína , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/genética , Sinapsis/fisiología
6.
Nat Struct Mol Biol ; 12(8): 663-70, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16025128

RESUMEN

Cells have evolved sophisticated DNA repair systems to correct damaged DNA. However, the human DNA mismatch repair protein Msh2-Msh3 is involved in the process of trinucleotide (CNG) DNA expansion rather than repair. Using purified protein and synthetic DNA substrates, we show that Msh2-Msh3 binds to CAG-hairpin DNA, a prime candidate for an expansion intermediate. CAG-hairpin binding inhibits the ATPase activity of Msh2-Msh3 and alters both nucleotide (ADP and ATP) affinity and binding interfaces between protein and DNA. These changes in Msh2-Msh3 function depend on the presence of A.A mispaired bases in the stem of the hairpin and on the hairpin DNA structure per se. These studies identify critical functional defects in the Msh2-Msh3-CAG hairpin complex that could misdirect the DNA repair process.


Asunto(s)
Disparidad de Par Base/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Modelos Genéticos , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Expansión de Repetición de Trinucleótido/genética , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Emparejamiento Base , Secuencia de Bases , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteína 2 Homóloga a MutS , Proteína 3 Homóloga de MutS , Unión Proteica , Proteínas/genética , Proteínas Proto-Oncogénicas/genética
7.
Cell Rep ; 32(9): 108104, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877683

RESUMEN

Rare variants in GRIN genes, which encode NMDAR subunits, are strongly associated with neurodevelopmental disorders. Among these, GRIN2A, which encodes the GluN2A subunit of NMDARs, is widely accepted as an epilepsy-causative gene. Here, we functionally characterize the de novo GluN2A-S1459G mutation identified in an epilepsy patient. We show that S1459 is a CaMKIIα phosphorylation site, and that endogenous phosphorylation is regulated during development and in response to synaptic activity in a dark rearing model. GluN2A-S1459 phosphorylation results in preferential binding of NMDARs to SNX27 and a corresponding decrease in PSD-95 binding, which consequently regulates NMDAR trafficking. Furthermore, the epilepsy-associated GluN2A-S1459G variant displays defects in interactions with both SNX27 and PSD-95, resulting in trafficking deficits, reduced spine density, and decreased excitatory synaptic transmission. These data demonstrate a role for CaMKIIα phosphorylation of GluN2A in receptor targeting and implicate NMDAR trafficking defects as a link to epilepsy.


Asunto(s)
Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Epilepsia/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Epilepsia/genética , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética
8.
Neuron ; 106(5): 759-768.e7, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32243781

RESUMEN

Autism spectrum disorder (ASD) is more prevalent in males; however, the etiology for this sex bias is not well understood. Many mutations on X-linked cell adhesion molecule NLGN4X result in ASD or intellectual disability. NLGN4X is part of an X-Y pair, with NLGN4Y sharing ∼97% sequence homology. Using biochemistry, electrophysiology, and imaging, we show that NLGN4Y displays severe deficits in maturation, surface expression, and synaptogenesis regulated by one amino acid difference with NLGN4X. Furthermore, we identify a cluster of ASD-associated mutations surrounding the critical amino acid in NLGN4X, and these mutations phenocopy NLGN4Y. We show that NLGN4Y cannot compensate for the functional deficits observed in ASD-associated NLGN4X mutations. Altogether, our data reveal a potential pathogenic mechanism for male bias in NLGN4X-associated ASD.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Cromosomas Humanos X/genética , Cromosomas Humanos Y/genética , Neuronas/metabolismo , Trastorno del Espectro Autista/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual/genética , Masculino , Mutación , Transporte de Proteínas/genética
9.
Cell Rep ; 29(10): 2944-2952.e5, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31801062

RESUMEN

The RhoGEFs Kalirin-7 and Trio are regulators of synaptic plasticity, and their dysregulation is associated with a range of neurodevelopmental and neurodegenerative disorders. Although studies have implicated both Kalirin and Trio in certain diseases, such as tauopathies, they remarkably differ in their association with other disorders. Using unbiased proteomics, we identified interactomes of Kalirin-7 and Trio to ascertain distinct protein association networks associated with their respective function and revealed groups of proteins that preferentially interact with a particular RhoGEF. In comparison, we find Trio interacts with a range of axon guidance and presynaptic complexes, whereas Kalirin-7 associates with several synaptic adhesion molecules. Specifically, we show Kalirin-7 is an interactor of the cell adhesion molecule neuroligin-1 (NLGN1), and NLGN1-dependent synaptic function is mediated through Kalirin-7 in an interaction-dependent manner. Our data reveal not only the interactomes of two important disease-related proteins, but also provide an intracellular effector of NLGN1 function.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas Sprague-Dawley , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Sinapsis/metabolismo
10.
Mol Cell Biol ; 24(18): 8195-209, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15340079

RESUMEN

Recent data in invertebrates demonstrated that huntingtin (htt) is essential for fast axonal trafficking. Here, we provide direct and functional evidence that htt is involved in fast axonal trafficking in mammals. Moreover, expression of full-length mutant htt (mhtt) impairs vesicular and mitochondrial trafficking in mammalian neurons in vitro and in whole animals in vivo. Particularly, mitochondria become progressively immobilized and stop more frequently in neurons from transgenic animals. These defects occurred early in development prior to the onset of measurable neurological or mitochondrial abnormalities. Consistent with a progressive loss of function, wild-type htt, trafficking motors, and mitochondrial components were selectively sequestered by mhtt in human Huntington's disease-affected brain. Data provide a model for how loss of htt function causes toxicity; mhtt-mediated aggregation sequesters htt and components of trafficking machinery leading to loss of mitochondrial motility and eventual mitochondrial dysfunction.


Asunto(s)
Transporte Axonal/fisiología , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Transporte Axonal/genética , Secuencia de Bases , Encéfalo/metabolismo , ADN/genética , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Técnicas In Vitro , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Neurológicos , Movimiento , Proteínas del Tejido Nervioso/deficiencia , Neuronas/ultraestructura , Proteínas Nucleares/deficiencia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
11.
Nat Neurosci ; 17(1): 56-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24336150

RESUMEN

Neuroligins are postsynaptic cell adhesion molecules that are important for synaptic function through their trans-synaptic interaction with neurexins (NRXNs). The localization and synaptic effects of neuroligin-1 (NL-1, also called NLGN1) are specific to excitatory synapses with the capacity to enhance excitatory synapses dependent on synaptic activity or Ca(2+)/calmodulin kinase II (CaMKII). Here we report that CaMKII robustly phosphorylates the intracellular domain of NL-1. We show that T739 is the dominant CaMKII site on NL-1 and is phosphorylated in response to synaptic activity in cultured rodent neurons and sensory experience in vivo. Furthermore, a phosphodeficient mutant (NL-1 T739A) reduces the basal and activity-driven surface expression of NL-1, leading to a reduction in neuroligin-mediated excitatory synaptic potentiation. To the best of our knowledge, our results are the first to demonstrate a direct functional interaction between CaMKII and NL-1, two primary components of excitatory synapses.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Bencilaminas/farmacología , Bicuculina/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/genética , Femenino , Antagonistas del GABA/farmacología , Regulación de la Expresión Génica/genética , Guanilato-Quinasas/metabolismo , Hipocampo/citología , Humanos , Inmunoprecipitación , Técnicas In Vitro , Proteínas Luminiscentes/genética , Masculino , Espectrometría de Masas , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/farmacología , Mutación/genética , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fosforilación/genética , Inhibidores de Proteínas Quinasas/farmacología , Receptores AMPA/genética , Privación Sensorial/fisiología , Análisis de Secuencia de Proteína , Estadísticas no Paramétricas , Sulfonamidas/farmacología , Transfección , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Corteza Visual/metabolismo
12.
J Biol Chem ; 281(24): 16583-90, 2006 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-16606616

RESUMEN

NR2C-containing N-methyl-D-aspartate (NMDA) receptors are highly expressed in cerebellar granule cells where they mediate the majority of current in the adult. NMDA receptors composed of NR1/NR2C exhibit a low conductance and reduced sensitivity to Mg(2+), compared with the more commonly studied NR2A- and NR2B-containing receptors. Despite these interesting features, very little is known about the regulation of NR2C function. Here we investigate the role of phosphorylation of NR2C in regulating NMDA receptor trafficking and ion channel properties. We identify a phosphorylation site, serine 1244 (Ser(1244)), near the extreme COOH terminus of NR2C, which is phosphorylated by both cAMP-dependent protein kinase and protein kinase C. This residue is located adjacent to the consensus PDZ ligand, a region that regulates protein-protein interactions and receptor trafficking in NR2A and NR2B. We show that Ser(1244) on NR2C is phosphorylated in vitro, in heterologous cells, and in neurons. Moreover, we demonstrate for the first time that NR2C interacts with the PSD-95 family of PDZ domain-containing proteins but that phosphorylation of Ser(1244) does not influence this PDZ interaction. Furthermore, Ser(1244) phosphorylation does not regulate surface expression of NR1/NR2C receptors. However, we find that this site does regulate the kinetics of the ion channel: a phosphomimetic mutation at Ser(1244) accelerates both the rise and decay of NMDA-evoked currents in excised patches from HEK-293 cells. Therefore, phosphorylation of Ser(1244) does not regulate trafficking but unexpectedly affects ion channel function, suggesting that phosphorylation of Ser(1244) on NR2C may be important in defining the functional properties of NMDA receptor-mediated currents in the cerebellum.


Asunto(s)
Regulación de la Expresión Génica , Receptores de N-Metil-D-Aspartato/química , Secuencia de Aminoácidos , Animales , Separación Celular , Cerebelo/metabolismo , Citometría de Flujo , Humanos , Datos de Secuencia Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Serina/química
13.
Proc Natl Acad Sci U S A ; 100(21): 12171-6, 2003 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-14527999

RESUMEN

There has been a longstanding debate regarding the role of proteolysis in Huntington's disease. The toxic peptide theory posits that N-terminal cleavage fragments of mutant Huntington's disease protein [mutant huntingtin (mhtt)] enter the nucleus to cause transcriptional dysfunction. However, recent data suggest a second model in which proteolysis of full-length mhtt is inhibited. Importantly, the two competing theories differ with respect to subcellular distribution of mhtt at initiation of toxicity: nuclear if cleaved and cytoplasmic in the absence of cleavage. Using quantitative single-cell analysis and time-lapse imaging, we show here that transcriptional dysfunction is "downstream" of cytoplasmic dysfunction. Primary and reversible toxic events involve destabilization of microtubules mediated by full-length mhtt before cleavage. Restoration of microtubule structure by taxol inhibits nuclear entry and increases cell survival.


Asunto(s)
Enfermedad de Huntington/etiología , Enfermedad de Huntington/metabolismo , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Muerte Celular , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoplasma/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Microtúbulos/efectos de los fármacos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/toxicidad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/genética , Proteínas Nucleares/toxicidad , Paclitaxel/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA