Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Biol Chem ; 288(38): 27396-27405, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23943618

RESUMEN

Down-regulation of protein phosphatase 2A (PP2A) methylation occurs in Alzheimer disease (AD). However, the regulation of PP2A methylation remains poorly understood. We have reported that altered leucine carboxyl methyltransferase (LCMT1)-dependent PP2A methylation is associated with down-regulation of PP2A holoenzymes containing the Bα subunit (PP2A/Bα) and subsequent accumulation of phosphorylated Tau in N2a cells, in vivo and in AD. Here, we show that pools of LCMT1, methylated PP2A, and PP2A/Bα are co-enriched in cholesterol-rich plasma membrane microdomains/rafts purified from N2a cells. In contrast, demethylated PP2A is preferentially distributed in non-rafts wherein small amounts of the PP2A methylesterase PME-1 are exclusively present. A methylation-incompetent PP2A mutant is excluded from rafts. Enhanced methylation of PP2A promotes the association of PP2A and Tau with the plasma membrane. Altered PP2A methylation following expression of a catalytically inactive LCMT1 mutant, knockdown of LCMT1, or alterations in one-carbon metabolism all result in a loss of plasma membrane-associated PP2A and Tau in N2a cells. This correlates with accumulation of soluble phosphorylated Tau, a hallmark of AD and other tauopathies. Thus, our findings reveal a distinct compartmentalization of PP2A and PP2A regulatory enzymes in plasma membrane microdomains and identify a novel methylation-dependent mechanism involved in modulating the targeting of PP2A, and its substrate Tau, to the plasma membrane. We propose that alterations in the membrane localization of PP2A and Tau following down-regulation of LCMT1 may lead to PP2A and Tau dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Regulación Enzimológica de la Expresión Génica , Microdominios de Membrana/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Línea Celular Tumoral , Colesterol/genética , Colesterol/metabolismo , Regulación hacia Abajo/genética , Técnicas de Silenciamiento del Gen , Humanos , Microdominios de Membrana/genética , Microdominios de Membrana/patología , Metilación , Fosforilación/genética , Proteína O-Metiltransferasa/biosíntesis , Proteína O-Metiltransferasa/genética , Proteína Fosfatasa 2/genética , Transporte de Proteínas/genética , Proteínas tau/genética
2.
J Neurosci ; 32(27): 9173-81, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22764226

RESUMEN

Folate deficiency and hypomethylation have been implicated in a number of age-related neurodegenerative disorders including dementia and Parkinson's disease (PD). Levodopa (L-dopa) therapy in PD patients has been shown to cause an increase in plasma total homocysteine as well as depleting cellular concentrations of the methyl donor, S-adenosylmethionine (SAM), and increasing the demethylated product S-adenosylhomocysteine (SAH). Modulation of the cellular SAM/SAH ratio can influence activity of methyltransferase enzymes, including leucine carboxyl methyltransferase that specifically methylates Ser/Thr protein phosphatase 2A (PP2A), a major Tau phosphatase. Here we show in human SH-SY5Y cells, in dopaminergic neurons, and in wild-type mice that l-dopa results in a reduced SAM/SAH ratio that is associated with hypomethylation of PP2A and increased phosphorylation of Tau (p-Tau) at the Alzheimer's disease-like PHF-1 phospho-epitope. The effect of L-dopa on PP2A and p-Tau was exacerbated in cells exposed to folate deficiency. In the folate-deficient mouse model, L-dopa resulted in a marked depletion of SAM and an increase in SAH in various brain regions with parallel downregulation of PP2A methylation and increased Tau phosphorylation. L-Dopa also enhanced demethylated PP2A amounts in the liver. These findings reveal a novel mechanism involving methylation-dependent pathways in L-dopa induces PP2A hypomethylation and increases Tau phosphorylation, which may be potentially detrimental to neuronal cells.


Asunto(s)
Encéfalo/metabolismo , Neuronas Dopaminérgicas/enzimología , Levodopa/toxicidad , Degeneración Nerviosa/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas tau/metabolismo , Animales , Antiparkinsonianos/toxicidad , Encéfalo/efectos de los fármacos , Encéfalo/patología , Línea Celular Tumoral , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos , Masculino , Metilación/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/fisiopatología , Neuroblastoma , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Cultivo Primario de Células , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteínas tau/agonistas , Proteínas tau/biosíntesis
3.
J Biol Chem ; 287(18): 14984-93, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22403409

RESUMEN

The predominant brain microtubule-associated proteins MAP2 and tau play a critical role in microtubule cytoskeletal organization and function. We have previously reported that PP2A/Bα, a major protein phosphatase 2A (PP2A) holoenzyme, binds to and dephosphorylates tau, and regulates microtubule stability. Here, we provide evidence that MAP2 co-purifies with and is dephosphorylated by endogenous PP2A/Bα in bovine gray matter. It co-localizes with PP2A/Bα in immature and mature human neuronal cell bodies. PP2A co-immunoprecipitates with and directly interacts with MAP2. Using in vitro binding assays, we show that PP2A/Bα binds to MAP2c isoforms through a region encompassing the microtubule-binding domain and upstream proline-rich region. Tau and MAP2 compete for binding to and dephosphorylation by PP2A/Bα. Remarkably, the protein-tyrosine kinase Fyn, which binds to the proline-rich RTPPKSP motif conserved in both MAP2 and tau, inhibits the interaction of PP2A/Bα with either tau or MAP2c. The corresponding synthetic RTPPKSP peptide, but not the phosphorylated RpTPPKSP version, competes with Tau and MAP2c for binding to PP2A/Bα. Significantly, down-regulation of PP2A/Bα and deregulation of Fyn-Tau protein interactions have been linked to enhanced tau phosphorylation in Alzheimer disease. Together, our results suggest that PP2A/Bα is part of segregated MAP2 and tau signaling scaffolds that can coordinate the action of key kinases and phosphatases involved in modulating neuronal plasticity. Deregulation of these compartmentalized multifunctional protein complexes is likely to contribute to tau deregulation, microtubule disruption, and altered signaling in tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Secuencias de Aminoácidos , Animales , Bovinos , Línea Celular , Neuronas/patología , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos
4.
J Neurochem ; 115(6): 1455-65, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21044074

RESUMEN

Neuritic alterations are a major feature of many neurodegenerative disorders. Methylation of protein phosphatase 2A (PP2A) catalytic C subunit by the leucine carboxyl methyltransferase (LCMT1), and demethylation by the protein phosphatase methylesterase 1, is a critical PP2A regulatory mechanism. It modulates the formation of PP2A holoenzymes containing the Bα subunit, which dephosphorylate key neuronal cytoskeletal proteins, including tau. Significantly, we have reported that LCMT1, methylated C and Bα expression levels are down-regulated in Alzheimer disease-affected brain regions. In this study, we show that enhanced expression of LCMT1 in cultured N2a neuroblastoma cells, which increases endogenous methylated C and Bα levels, induces changes in F-actin organization. It promotes serum-independent neuritogenesis and development of extended tau-positive processes upon N2a cell differentiation. These stimulatory effects can be abrogated by LCMT1 knockdown and S-adenosylhomocysteine, an inhibitor of methylation reactions. Expression of protein phosphatase methylesterase 1 and the methylation-site L309Δ C subunit mutant, which decrease intracellular methylated C and Bα levels, block N2a cell differentiation and LCMT1-mediated neurite formation. Lastly, inducible and non-inducible knockdown of Bα in N2a cells inhibit process outgrowth. Altogether, our results establish a novel mechanistic link between PP2A methylation and development of neurite-like processes.


Asunto(s)
Hidrolasas de Éster Carboxílico/fisiología , Diferenciación Celular/fisiología , Metilación de ADN/fisiología , Neuroblastoma/enzimología , Neuroblastoma/patología , Proteína Fosfatasa 2/fisiología , Animales , Dominio Catalítico/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Metilación de ADN/genética , Técnicas de Silenciamiento del Gen , Ratones , Neuritas/enzimología , Neuritas/patología , Neuroblastoma/genética , Proteína O-Metiltransferasa/fisiología
5.
J Neurosci ; 28(45): 11477-87, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18987184

RESUMEN

Altered folate homeostasis is associated with many clinical and pathological manifestations in the CNS. Notably, folate-mediated one-carbon metabolism is essential for methyltransferase-dependent cellular methylation reactions. Biogenesis of protein phosphatase 2A (PP2A) holoenzyme containing the regulatory B(alpha) subunit, a major brain tau phosphatase, is controlled by methylation. Here, we show that folate deprivation in neuroblastoma cells induces downregulation of PP2A leucine carboxyl methyltransferase-1 (LCMT-1) expression, resulting in progressive accumulation of newly synthesized demethylated PP2A pools, concomitant loss of B(alpha), and ultimately cell death. These effects are further accentuated by overexpression of PP2A methylesterase (PME-1) but cannot be rescued by PME-1 knockdown. Overexpression of either LCMT-1 or B(alpha) is sufficient to protect cells against the accumulation of demethylated PP2A, increased tau phosphorylation, and cell death induced by folate starvation. Conversely, knockdown of either protein accelerates folate deficiency-evoked cell toxicity. Significantly, mice maintained for 2 months on low-folate or folate-deficient diets have brain-region-specific alterations in metabolites of the methylation pathway. Those are associated with downregulation of LCMT-1, methylated PP2A, and B(alpha) expression and enhanced tau phosphorylation in susceptible brain regions. Our studies provide novel mechanistic insights into the regulation of PP2A methylation and tau. They establish LCMT-1- and B(alpha)-containing PP2A holoenzymes as key mediators of the role of folate in the brain. Our results suggest that counteracting the neuronal loss of LCMT-1 and B(alpha) could be beneficial for all tauopathies and folate-dependent disorders of the CNS.


Asunto(s)
Encéfalo/metabolismo , Regulación hacia Abajo/fisiología , Deficiencia de Ácido Fólico/metabolismo , Proteína O-Metiltransferasa/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/patología , Hidrolasas de Éster Carboxílico/fisiología , Muerte Celular/fisiología , Línea Celular Transformada , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Serina/metabolismo
6.
J Cell Biol ; 158(5): 967-78, 2002 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-12196510

RESUMEN

Tight junctions (TJs) play a crucial role in the establishment of cell polarity and regulation of paracellular permeability in epithelia. Here, we show that upon calcium-induced junction biogenesis in Madin-Darby canine kidney cells, ABalphaC, a major protein phosphatase (PP)2A holoenzyme, is recruited to the apical membrane where it interacts with the TJ complex. Enhanced PP2A activity induces dephosphorylation of the TJ proteins, ZO-1, occludin, and claudin-1, and is associated with increased paracellular permeability. Expression of PP2A catalytic subunit severely prevents TJ assembly. Conversely, inhibition of PP2A by okadaic acid promotes the phosphorylation and recruitment of ZO-1, occludin, and claudin-1 to the TJ during junctional biogenesis. PP2A negatively regulates TJ assembly without appreciably affecting the organization of F-actin and E-cadherin. Significantly, inhibition of atypical PKC (aPKC) blocks the calcium- and serum-independent membrane redistribution of TJ proteins induced by okadaic acid. Indeed, PP2A associates with and critically regulates the activity and distribution of aPKC during TJ formation. Thus, we provide the first evidence for calcium-dependent targeting of PP2A in epithelial cells, we identify PP2A as the first serine/threonine phosphatase associated with the multiprotein TJ complex, and we unveil a novel role for PP2A in the regulation of epithelial aPKC and TJ assembly and function.


Asunto(s)
Epitelio/enzimología , Fosfoproteínas Fosfatasas/metabolismo , Proteína Quinasa C/metabolismo , Uniones Estrechas/enzimología , Actinas/metabolismo , Animales , Calcio/metabolismo , Adhesión Celular , Línea Celular , Colon/citología , Perros , Epitelio/ultraestructura , Humanos , Microscopía Electrónica , Permeabilidad , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosforilación , Unión Proteica , Proteína Fosfatasa 2
7.
J Neurosci ; 27(11): 2751-9, 2007 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-17360897

RESUMEN

Alzheimer's disease (AD) neuropathology is characterized by the accumulation of phosphorylated tau and amyloid-beta peptides derived from the amyloid precursor protein (APP). Elevated blood levels of homocysteine are a significant risk factor for many age-related diseases, including AD. Impaired homocysteine metabolism favors the formation of S-adenosylhomocysteine, leading to inhibition of methyltransferase-dependent reactions. Here, we show that incubation of neuroblastoma cells with S-adenosylhomocysteine results in reduced methylation of protein phosphatase 2A (PP2A), a major brain Ser/Thr phosphatase, most likely by inhibiting PP2A methyltransferase (PPMT). PP2A methylation levels are also decreased after ectopic expression of PP2A methylesterase in Neuro-2a (N2a) cells. Reduced PP2A methylation promotes the downregulation of B alpha-containing holoenzymes, thereby affecting PP2A substrate specificity. It is associated with the accumulation of both phosphorylated tau and APP isoforms and increased secretion of beta-secretase-cleaved APP fragments and amyloid-beta peptides. Conversely, incubation of N2a cells with S-adenosylmethionine and expression of PPMT enhance PP2A methylation. This leads to the accumulation of dephosphorylated tau and APP species and increased secretion of neuroprotective alpha-secretase-cleaved APP fragments. Remarkably, hyperhomocysteinemia induced in wild-type and cystathionine-beta-synthase +/- mice by feeding a high-methionine, low-folate diet is associated with increased brain S-adenosylhomocysteine levels, PPMT downregulation, reduced PP2A methylation levels, and tau and APP phosphorylation. We reported previously that downregulation of neuronal PPMT and PP2A methylation occur in affected brain regions from AD patients. The link between homocysteine, PPMT, PP2A methylation, and key CNS proteins involved in AD pathogenesis provides new mechanistic insights into this disorder.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Homocisteína/metabolismo , Metiltransferasas/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas tau/fisiología , Animales , Línea Celular Tumoral , Homocisteína/genética , Metilación , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 2
8.
J Neurochem ; 101(4): 959-71, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17394530

RESUMEN

Carboxymethylation and phosphorylation of protein phosphatase 2A (PP2A) catalytic C subunit are evolutionary conserved mechanisms that critically control PP2A holoenzyme assembly and substrate specificity. Down-regulation of PP2A methylation and PP2A enzymes containing the B alpha regulatory subunit occur in Alzheimer's disease. In this study, we show that expressed wild-type and methylation- (L309 Delta) and phosphorylation- (T304D, T304A, Y307F, and Y307E) site mutants of PP2A C subunit differentially bind to B, B', and B''-type regulatory subunits in NIH 3T3 fibroblasts and neuro-2a (N2a) neuroblastoma cells. They also display distinct binding affinity for microtubules (MTs). Relative to controls, expression of the wild-type, T304A and Y307F C subunits in N2a cells promotes the accumulation of acetylated and detyrosinated MTs. However, expression of the Y307E, L309 Delta, and T304D mutants, which are impaired in their ability to associate with the B alpha subunit, induces their loss. Silencing of B alpha subunit in N2a and NIH 3T3 cells is sufficient to induce a similar breakdown of acetylated and detyrosinated MTs. It also confers increased sensitivity to nocodazole-induced MT depolymerization. Our findings suggest that changes in intracellular PP2A subunit composition can modulate MT dynamics. They support the hypothesis that reduced amounts of neuronal B alpha-containing PP2A heterotrimers contribute to MT destabilization in Alzheimer's disease.


Asunto(s)
Regulación de la Expresión Génica/genética , Microtúbulos/metabolismo , Mutación/fisiología , Fosfoproteínas Fosfatasas/metabolismo , Acetilación , Animales , Línea Celular , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Neuroblastoma , Ácido Ocadaico/farmacología , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 2 , Interferencia de ARN/fisiología , Transfección/métodos , Tirosina/metabolismo
9.
J Virol ; 77(5): 2807-18, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12584304

RESUMEN

There is increasing evidence that the transforming DNA tumor virus simian virus 40 (SV40) is associated with human malignancies. SV40 small tumor antigen (small t) interacts with endogenous serine/threonine protein phosphatase 2A (PP2A) and is required for the transforming activity of SV40 in epithelial cells of the lung and kidney. Here, we show that expression of SV40 small t in epithelial MDCK cells induces acute morphological changes and multilayering. Significantly, it also causes severe defects in the biogenesis and barrier properties of tight junctions (TJs) but does not prevent formation of adherens junctions. Small t-induced TJ defects are associated with a loss of PP2A from areas of cell-cell contact; altered distribution and reduced amounts of the TJ proteins ZO-1, occludin, and claudin-1; and marked disorganization of the actin cytoskeleton. Small t-mediated F-actin rearrangements encompass increased Rac-induced membrane ruffling and lamellipodia, Cdc42-initiated filopodia, and loss of Rho-dependent stress fibers. Indeed, these F-actin changes coincide with elevated levels of Rac1 and Cdc42 and decreased amounts of RhoA in small t-expressing cells. Notably, these cellular effects of small t are dependent on its interaction with endogenous PP2A. Thus, our findings provide the first evidence that, in polarized epithelial cells, expression of small t alone is sufficient to induce deregulation of Rho GTPases, F-actin, and intercellular adhesion, through interaction with endogenous PP2A. Because defects in the actin cytoskeleton and TJ disruption have been linked to loss of cell polarity and tumor invasiveness, their deregulation by PP2A and small t likely contributes to the role of SV40 in epithelial cell transformation.


Asunto(s)
Antígenos Transformadores de Poliomavirus/metabolismo , Transformación Celular Viral , Citoesqueleto/ultraestructura , Uniones Estrechas/ultraestructura , Actinas/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/genética , Línea Celular , Citoesqueleto/metabolismo , Perros , Células Epiteliales , Humanos , Riñón/citología , Microscopía Electrónica , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2 , Uniones Estrechas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA