Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 4(1): 658, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079065

RESUMEN

It has not been well studied which cells and related mechanisms contribute to endochondral ossification. Here, we fate mapped the leptin receptor-expressing (LepR+) mesenchymal stem cells (MSCs) in different embryonic and adult extremities using Lepr-cre; tdTomato mice and investigated the underling mechanism using Lepr-cre; Ppp2r1afl/fl mice. Tomato+ cells appear in the primary and secondary ossification centers and express the hypertrophic markers. Ppp2r1a deletion in LepR+ MSCs reduces the expression of Runx2, Osterix, alkaline phosphatase, collagen X, and MMP13, but increases that of the mature adipocyte marker perilipin, thereby reducing trabecular bone density and enhancing fat content. Mechanistically, PP2A dephosphorylates Runx2 and BRD4, thereby playing a major role in positively and negatively regulating osteogenesis and adipogenesis, respectively. Our data identify LepR+ MSC as the cell origin of endochondral ossification during embryonic and postnatal bone growth and suggest that PP2A is a therapeutic target in the treatment of dysregulated bone formation.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Proteína Fosfatasa 2/metabolismo , Receptores de Leptina/metabolismo , Adipogénesis , Animales , Densidad Ósea , Huesos/citología , Huesos/embriología , Huesos/metabolismo , Diferenciación Celular , Proliferación Celular , Condrogénesis , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/metabolismo , Fosforilación , Embarazo , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Factores de Transcripción/metabolismo
2.
Gynecol Oncol ; 162(1): 182-189, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33867147

RESUMEN

OBJECTIVE: PI3K-AKT pathway mutations initiate a kinase cascade that characterizes endometrial cancer (EC). As kinases seldom cause oncogenic transformation without dysregulation of antagonistic phosphatases, pivotal interactions governing this pathway were explored and correlated with clinical outcomes. METHODS: After exclusion of patients with POLE mutations from The Cancer Genome Atlas EC cohort with endometrioid or serous EC, the study population was 209 patients with DNA sequencing, quantitative gene-specific RNA expression, copy number variation (CNV), and surveillance data available. Extracted data were annotated and integrated. RESULTS: A PIK3CA, PTEN, or PIK3R1 mutant (-mu) was present in 83% of patients; 57% harbored more than 1 mutation without adversely impacting progression-free survival (PFS) (P = .10). PIK3CA CNV of at least 1.1 (CNV high [-H]) was detected in 26% and linked to TP53-mu and CIP2A expression (P < .001) but was not associated with PFS (P = .24). PIK3CA expression was significantly different between those with CIP2A-H and CIP2A low (-L) expression (the endogenous inhibitor of protein phosphatase 2A [PP2A]), when stratified by PIK3CA mutational status or by PIK3CA CNV-H and CNV-L (all P < .01). CIP2A-H or PPP2R1A-mu mitigates PP2A kinase dephosphorylation, and FBXW7-mu nullifies E3 ubiquitin ligase (E3UL) oncoprotein degradation. CIP2A-H and PPP2R1A-mu (PP2A impairment) and FBXW7-mu (E3UL impairment) were associated with compromised PFS (P < .001) and were prognostically discriminatory for PIK3CA-mu and PIK3CA CNV-H tumors (P < .001). Among documented recurrences, 84% were associated with impaired PP2A (75%) and/or E3UL (20%). CONCLUSION: PP2A and E3UL deficiencies are seminal biological drivers in EC independent of PIK3CA-mu, PTEN-mu, and PIK3R1-mu and PIK3CA CNV.


Asunto(s)
Neoplasias Endometriales/enzimología , Proteína Fosfatasa 2/deficiencia , Ubiquitina-Proteína Ligasas/deficiencia , Neoplasias Abdominales , Autoantígenos/biosíntesis , Autoantígenos/genética , Fosfatidilinositol 3-Quinasa Clase I/biosíntesis , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase Ia/biosíntesis , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Mutación , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Cancer Res ; 80(16): 3305-3318, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32522823

RESUMEN

There is currently a lack of precise predictive biomarkers for patient selection in clinical trials of inhibitors targeting replication stress (RS) response proteins ATR and CHK1. The objective of this study was to identify novel predictive biomarkers for the response to these agents in treating non-small cell lung cancer (NSCLC). A genome-wide loss-of-function screen revealed that tumor suppressor PPP2R2A, a B regulatory subunit of protein phosphatase 2 (PP2A), determines sensitivity to CHK1 inhibition. A synthetic lethal interaction between PPP2R2A deficiency and ATR or CHK1 inhibition was observed in NSCLC in vitro and in vivo and was independent of p53 status. ATR and CHK1 inhibition resulted in significantly increased levels of RS and altered replication dynamics, particularly in PPP2R2A-deficient NSCLC cells. Mechanistically, PPP2R2A negatively regulated translation of oncogene c-Myc protein. c-Myc activity was required for PPP2R2A deficiency-induced alterations of replication initiation/RS and sensitivity to ATR/CHK1 inhibitors. We conclude that PPP2R2A deficiency elevates RS by upregulating c-Myc activity, rendering cells reliant on the ATR/CHK1 axis for survival. Our studies show a novel synthetic lethal interaction and identify PPP2R2A as a potential new predictive biomarker for patient stratification in the clinical use of ATR and CHK1 inhibitors. SIGNIFICANCE: This study reveals new approaches to specifically target PPP2R2A-deficient lung cancer cells and provides a novel biomarker that will significantly improve treatment outcome with ATR and CHK1 inhibitors.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Biomarcadores de Tumor/deficiencia , Carcinoma de Pulmón de Células no Pequeñas/química , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Neoplasias Pulmonares/química , Proteína Fosfatasa 2/deficiencia , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Daño del ADN , Replicación del ADN , Resistencia a Antineoplásicos , Femenino , Técnicas de Silenciamiento del Gen , Genes p53 , Estudio de Asociación del Genoma Completo , Xenoinjertos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Desnudos , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño
4.
Cell Commun Signal ; 17(1): 91, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31387591

RESUMEN

BACKGROUND: Protein phosphorylation & dephosphorylation are ubiquitous cellular processes that allow for the nuanced and reversible regulation of protein activity. Protein phosphatase 2A (PP2A) is a multifunction phosphatase that is well expressed in all cell types of kidney during early renal development, though its functions in kidney remains to be elucidated. METHODS: PP2A conditional knock-out mice was generated with PP2A fl/fl mice that were crossed with Podocin-Cre mice. The phenotype of Pod-PP2A-KO mice (homozygous for the floxed PP2A allele with Podocin-Cre) and littermate PP2A fl/fl controls (homozygous for the PP2A allele but lacking Podocin-Cre) were further studied. Primary podocytes isolated from the Pod-PP2A-KO mice were cultured and they were then employed with sing label-free nano-LC - MS/MS technology on a Q-exactive followed by SIEVE processing to identify possible target molecular entities for the dephosphorylation effect of PP2A, in which Western blot and immunofluorescent staining were used to analyze further. RESULTS: Pod-PP2A-KO mice were developed with weight loss, growth retardation, proteinuria, glomerulopathy and foot process effacement, together with reduced expression of some slit diaphragm molecules and cytoskeleton rearrangement of podocytes. Y box protein 1 (YB-1) was identified to be the target molecule for dephosphorylation effect of PP2A. Furthermore, YB-1 phosphorylation was up-regulated in the Pod-PP2A-KO mice in contrast to the wild type controls, while total and un-phosphorylated YB-1 both was moderately down-regulated in podocytes from the Pod-PP2A-KO mice. CONCLUSION: Our study revealed the important role of PP2A in regulating the development of foot processes and fully differentiated podocytes whereas fine-tuning of YB-1 via a post-translational modification by PP2A regulating its activity might be crucial for the functional integrity of podocytes and glomerular filtration barrier.


Asunto(s)
Glomérulos Renales/citología , Glomérulos Renales/fisiología , Podocitos/citología , Proteína Fosfatasa 2/metabolismo , Animales , Peso Corporal , Citoesqueleto/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Masculino , Ratones , Fosforilación , Podocitos/patología , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Proteinuria/enzimología , Proteinuria/genética , Proteína 1 de Unión a la Caja Y/metabolismo
5.
Mol Brain ; 12(1): 51, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113458

RESUMEN

Protein phosphorylation plays an important role in learning and memory. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in the regulation of neural synaptic plasticity. Here, to determine if PP2A is necessary for successful learning and memory, we have utilized a Tg (Camk2a-cre) T29-2Stl mice to specific knock down the expression of hippocampal PP2A in mice. By analysing behavioural, we observed that loss of PP2A in the hippocampal CA1 area did not affect the formation of memory but impaired contextual fear memory extinction. We use the electrophysiological recording to find the synaptic mechanisms. The results showed that the basic synapse transmission and synaptic plasticity of PP2A conditional knockout (CKO) mice were impaired. Moreover, PP2A CKO mice exhibited a saturating long-term potentiation inducted by strong theta burst stimulation but no depotentiation after low-frequency stimulation. Taken together, our results provide the evidence that PP2A is involved in synaptic transmission and hippocampus-dependent memory extinction.


Asunto(s)
Región CA1 Hipocampal/enzimología , Región CA1 Hipocampal/fisiología , Extinción Psicológica , Memoria , Proteína Fosfatasa 2/deficiencia , Animales , Conducta Exploratoria , Femenino , Locomoción , Potenciación a Largo Plazo , Masculino , Ratones Noqueados , Plasticidad Neuronal , Proteína Fosfatasa 2/metabolismo , Transmisión Sináptica
6.
Circ Res ; 124(5): 737-746, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30602331

RESUMEN

RATIONALE: Voltage-gated Na+ channel ( INa) function is critical for normal cardiac excitability. However, the Na+ channel late component ( INa,L) is directly associated with potentially fatal forms of congenital and acquired human arrhythmia. CaMKII (Ca2+/calmodulin-dependent kinase II) enhances INa,L in response to increased adrenergic tone. However, the pathways that negatively regulate the CaMKII/Nav1.5 axis are unknown and essential for the design of new therapies to regulate the pathogenic INa,L. OBJECTIVE: To define phosphatase pathways that regulate INa,L in vivo. METHODS AND RESULTS: A mouse model lacking a key regulatory subunit (B56α) of the PP (protein phosphatase) 2A holoenzyme displayed aberrant action potentials after adrenergic stimulation. Unbiased computational modeling of B56α KO (knockout) mouse myocyte action potentials revealed an unexpected role of PP2A in INa,L regulation that was confirmed by direct INa,L recordings from B56α KO myocytes. Further, B56α KO myocytes display decreased sensitivity to isoproterenol-induced induction of arrhythmogenic INa,L, and reduced CaMKII-dependent phosphorylation of Nav1.5. At the molecular level, PP2A/B56α complex was found to localize and coimmunoprecipitate with the primary cardiac Nav channel, Nav1.5. CONCLUSIONS: PP2A regulates Nav1.5 activity in mouse cardiomyocytes. This regulation is critical for pathogenic Nav1.5 late current and requires PP2A-B56α. Our study supports B56α as a novel target for the treatment of arrhythmia.


Asunto(s)
Arritmias Cardíacas/enzimología , Frecuencia Cardíaca , Activación del Canal Iónico , Miocitos Cardíacos/enzimología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Proteína Fosfatasa 2/metabolismo , Potenciales de Acción , Agonistas Adrenérgicos beta/farmacología , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Humanos , Activación del Canal Iónico/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Fenotipo , Fosforilación , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Factores de Tiempo
7.
Cells ; 9(1)2019 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-31905853

RESUMEN

In most cancers, cellular origin and the contribution of intrinsic and extrinsic factors toward transformation remain elusive. Cell specific carcinogenesis models are currently unavailable. To investigate cellular origin in carcinogenesis, we developed a tumorigenesis model based on a combination of carcinogenesis and genetically engineered mouse models. We show in organoids that treatment of any of three carcinogens, DMBA, MNU, or PhIP, with protein phosphatase 2A (PP2A) knockout induced tumorigenesis in Lgr5+ intestinal lineage, but not in differentiated cells. These transformed cells increased in stem cell signature, were upregulated in EMT markers, and acquired tumorigenecity. A mechanistic approach demonstrated that tumorigenesis was dependent on Wnt, PI3K, and RAS-MAPK activation. In vivo combination with carcinogen and PP2A depletion also led to tumor formation. Using whole-exome sequencing, we demonstrate that these intestinal tumors display mutation landscape and core driver pathways resembling human intestinal tumor in The Cancer Genome Atlas (TCGA). These data provide a basis for understanding the interplay between extrinsic carcinogen and intrinsic genetic modification and suggest that PP2A functions as a tumor suppressor in intestine carcinogenesis.


Asunto(s)
Carcinogénesis/metabolismo , Intestinos/patología , Proteína Fosfatasa 2/deficiencia , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/patología , Animales , Carcinogénesis/patología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Transformación Celular Neoplásica , Femenino , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Organoides/metabolismo , Proteína Fosfatasa 2/metabolismo , Células Madre/citología , Vía de Señalización Wnt , beta Catenina/metabolismo
8.
Cardiovasc Res ; 115(3): 519-529, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30203051

RESUMEN

AIMS: B56α is a protein phosphatase 2A (PP2A) regulatory subunit that is highly expressed in the heart. We previously reported that cardiomyocyte B56α localizes to myofilaments under resting conditions and translocates to the cytosol in response to acute ß-adrenergic receptor (ß-AR) stimulation. Given the importance of reversible protein phosphorylation in modulating cardiac function during sympathetic stimulation, we hypothesized that loss of B56α in mice with targeted disruption of the gene encoding B56α (Ppp2r5a) would impact on cardiac responses to ß-AR stimulation in vivo. METHODS AND RESULTS: Cardiac phenotype of mice heterozygous (HET) or homozygous (HOM) for the disrupted Ppp2r5a allele and wild type (WT) littermates was characterized under basal conditions and following acute ß-AR stimulation with dobutamine (DOB; 0.75 mg/kg i.p.) or sustained ß-AR stimulation by 2-week infusion of isoproterenol (ISO; 30 mg/kg/day s.c.). Left ventricular (LV) wall thicknesses, chamber dimensions and function were assessed by echocardiography, and heart tissue collected for gravimetric, histological, and biochemical analyses. Western blot analysis revealed partial and complete loss of B56α protein in hearts from HET and HOM mice, respectively, and no changes in the expression of other PP2A regulatory, catalytic or scaffolding subunits. PP2A catalytic activity was reduced in hearts of both HET and HOM mice. There were no differences in the basal cardiac phenotype between genotypes. Acute DOB stimulation induced the expected inotropic response in WT and HET mice, which was attenuated in HOM mice. In contrast, DOB-induced increases in heart rate were unaffected by B56α deficiency. In WT mice, ISO infusion increased LV wall thicknesses, cardiomyocyte area and ventricular mass, without LV dilation, systolic dysfunction, collagen deposition or foetal gene expression. The hypertrophic response to ISO was blunted in mice deficient for B56α. CONCLUSION: These findings identify B56α as a potential regulator of cardiac structure and function during ß-AR stimulation.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Cardiomegalia/inducido químicamente , Dobutamina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Isoproterenol , Miocitos Cardíacos/efectos de los fármacos , Proteína Fosfatasa 2/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Cardiomegalia/enzimología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Homocigoto , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/enzimología , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética
9.
Cell ; 173(2): 470-484.e18, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29551267

RESUMEN

B cell activation during normal immune responses and oncogenic transformation impose increased metabolic demands on B cells and their ability to retain redox homeostasis. While the serine/threonine-protein phosphatase 2A (PP2A) was identified as a tumor suppressor in multiple types of cancer, our genetic studies revealed an essential role of PP2A in B cell tumors. Thereby, PP2A redirects glucose carbon utilization from glycolysis to the pentose phosphate pathway (PPP) to salvage oxidative stress. This unique vulnerability reflects constitutively low PPP activity in B cells and transcriptional repression of G6PD and other key PPP enzymes by the B cell transcription factors PAX5 and IKZF1. Reflecting B-cell-specific transcriptional PPP-repression, glucose carbon utilization in B cells is heavily skewed in favor of glycolysis resulting in lack of PPP-dependent antioxidant protection. These findings reveal a gatekeeper function of the PPP in a broad range of B cell malignancies that can be efficiently targeted by small molecule inhibition of PP2A and G6PD.


Asunto(s)
Carbono/metabolismo , Glucosa/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Glucólisis , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Estrés Oxidativo , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Vía de Pentosa Fosfato , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transcripción Genética
10.
Cell Biol Int ; 42(4): 403-410, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29068100

RESUMEN

Serine/threonine protein phosphatase 2A (PP2A) is involved in regulating various physiological processes including cell cycle, growth, apoptosis, and signal transduction. Osteoblast differentiation is controlled by main bone specific transcription factors including Osterix, distal-less homeobox 5 (Dlx5), and Runt-related transcription factor 2 (Runx2). We previously reported that knockdown of PP2A Cα increases the expression of Osterix, leading to the accelerated osteoblast differentiation through the upregulation of bone-related genes. In this study, we examined whether Dlx5 and Runx2 are involved in the upregulated Osterix expression in PP2A Cα-knockdown osteoblasts (shPP2A cells). The expression of Dlx5 as well as Osterix was significantly higher in shPP2A cells in the initial stage of osteoblast differentiation compared with the control cells (shCont). The expression of Runx2 protein was also higher in shPP2A cells compared with shCont cells although its mRNA level was comparable. Reduction of Dlx5 or Runx2 decreased Osterix expression and alkaline phosphatase activity in shPP2A cells. Luciferase assay showed that Osterix promoter activity was drastically elevated in shPP2A cells compared with that in shCont cells. The deletion or mutation of the Dlx5 and Runx2 binding sites significantly suppressed Osterix promoter activity in shPP2A cells. These results indicate that Dlx5 and Runx2 are critical factors for the upregulated Osterix expression in shPP2A cells, which is considered to be important for the accelerated osteoblast differentiation in these cells.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Homeodominio/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Proteína Fosfatasa 2/deficiencia , Factor de Transcripción Sp7/biosíntesis , Células 3T3 , Animales , Diferenciación Celular/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Ratones , Osteoblastos/enzimología , Osteogénesis , Regiones Promotoras Genéticas , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Regulación hacia Arriba
11.
Sci Rep ; 7(1): 1630, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28487562

RESUMEN

EG5 (KIF11) is a member of the kinesin-like protein family involved in centrosome separation and bipolar spindle formation. When a cell enters mitosis, CDK1 phosphorylates EG5 at Thr926 and promotes EG5 localization on the mitotic spindle which drives bipolar spindle formation. EG5 provides power for spindle movement and thus controls the dynamics of spindle assembly. However, little is known about EG5 regulation or how EG5 detaches from the spindle upon mitotic exit. In this study we identify EG5 as a novel substrate of PP2A phosphatase, and we show that the PP2A/B55α complex plays an important role in mitotic exit by a mechanism involving EG5. The PP2A/B55α complex physically associates with the EG5 C-terminal tail domain and dephosphorylates EG5 at Thr926 that enables mitotic exit. Conversely PP2A knockdown cells show a high level of phospho-EG5 in late metaphase, which is associated with a delay in mitotic exit. These phenotypic features are similar to those induced by EG5/T926D transfection that mimics phosphorylated EG5 status. Our results argue that PP2A controls mitotic exit through EG5 dephosphorylation. Lack of PP2A leads to abnormal EG5 activation, resulting in delay of mitotic exit.


Asunto(s)
Cinesinas/metabolismo , Mitosis , Proteína Fosfatasa 2/metabolismo , Anafase , Segregación Cromosómica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Metafase , Fosforilación , Fosfotreonina/metabolismo , Unión Proteica , Proteína Fosfatasa 2/deficiencia
12.
PLoS One ; 12(4): e0176538, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28448624

RESUMEN

Phosphate (Pi) limitation is a constraint for plant growth and development in many natural and agricultural ecosystems. In this study, a gene encoding Zea mays L. protein phosphatase 2A regulatory subunit A, designated ZmPP2AA1, was induced in roots by low Pi availability. The function of the ZmPP2AA1 gene in maize was analyzed using overexpression and RNA interference. ZmPP2AA1 modulated root gravitropism, negatively regulated primary root (PR) growth, and stimulated the development of lateral roots (LRs). A detailed characterization of the root system architecture (RSA) in response to different Pi concentrations with or without indole-3-acetic acid and 1-N-naphthylphthalamic acid revealed that auxin was involved in the RSA response to low Pi availability. Overexpression of ZmPP2AA1 enhanced tolerance to Pi starvation in transgenic maize in hydroponic and soil pot experiments. An increased dry weight (DW), root-to-shoot ratio, and total P content and concentration, along with a delayed and reduced accumulation of anthocyanin in overexpressing transgenic maize plants coincided with their highly branched root system and increased Pi uptake capability under low Pi conditions. Inflorescence development of the ZmPP2AA1 overexpressing line was less affected by low Pi stress, resulting in higher grain yield per plant under Pi deprivation. These data reveal the biological function of ZmPP2AA1, provide insights into a linkage between auxin and low Pi responses, and drive new strategies for the efficient utilization of Pi by maize.


Asunto(s)
Fosfatos/farmacología , Raíces de Plantas/crecimiento & desarrollo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Zea mays/crecimiento & desarrollo , Zea mays/genética , Secuencia de Aminoácidos , Clonación Molecular , Relación Dosis-Respuesta a Droga , Expresión Génica , Genómica , Filogenia , Raíces de Plantas/efectos de los fármacos , Plantas Modificadas Genéticamente , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/deficiencia , Interferencia de ARN , Análisis de Secuencia , Zea mays/efectos de los fármacos , Zea mays/metabolismo
13.
Cell Cycle ; 15(12): 1620-30, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27163342

RESUMEN

Sit4p is the catalytic subunit of a ceramide-activated PP2A-like phosphatase that regulates cell cycle, mitochondrial function, oxidative stress resistance and chronological lifespan in yeast. In this study, we show that hexokinase 2 (Hxk2p) is hyperphosphorylated in sit4Δ mutants grown in glucose medium by a Snf1p-independent mechanism and Hxk2p-S15A mutation suppresses phenotypes associated with SIT4 deletion, namely growth arrest at G1 phase, derepression of mitochondrial respiration, H2O2 resistance and lifespan extension. Consistently, the activation of Sit4p in isc1Δ mutants, which has been associated with premature aging, leads to Hxk2p hypophosphorylation, and the expression of Hxk2p-S15E increases the lifespan of isc1Δ cells. The overall results suggest that Hxk2p functions downstream of Sit4p in the control of cell cycle, mitochondrial function, oxidative stress resistance and chronological lifespan.


Asunto(s)
Regulación Fúngica de la Expresión Génica , Hexoquinasa/genética , Fosfoproteínas/genética , Proteína Fosfatasa 2/genética , Proteínas de Saccharomyces cerevisiae/genética , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Eliminación de Gen , Glucosa/metabolismo , Hexoquinasa/metabolismo , Peróxido de Hidrógeno/farmacología , Redes y Vías Metabólicas/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación , Consumo de Oxígeno , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/deficiencia , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal
14.
Biol Chem ; 397(6): 541-54, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26894577

RESUMEN

Mutations in the gene coding for the multi-domain protein leucine-rich repeat kinase 2 (LRRK2) are the leading cause of genetically inherited Parkinson's disease (PD). Two of the common found mutations are the R1441C and G2019S. In this study we identified protein phosphatase 2A (PP2A) as an interacting partner of LRRK2. We were able to demonstrate that the Ras of complex protein (ROC) domain is sufficient to interact with the three subunits of PP2A in human neuroblastoma SH-SY5Y cells and in HeLa cells. The alpha subunit of PP2A is interacting with LRRK2 in the perinuclear region of HeLa cells. Silencing the catalytic subunit of PP2A by shRNA aggravated cellular degeneration induced by the pathogenic R1441C-LRRK2 mutant expressed in neuroblastoma SH-SY5Y cells. A similar enhancement of apoptotic nuclei was observed by downregulation of the catalytic subunit of PP2A in cultured cortical cells derived from neurons overexpressing the pathogenic mutant G2019S-LRRK2. Conversely, pharmacological activation of PP2A by sodium selenate showed a partial neuroprotection from R1441C-LRRK2-induced cellular degeneration. All these data suggest that PP2A is a new interacting partner of LRRK2 and reveal the importance of PP2A as a potential therapeutic target in PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteína Fosfatasa 2/metabolismo , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/química , Neuronas/citología , Neuronas/efectos de los fármacos , Unión Proteica , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Ácido Selénico/farmacología
15.
FEBS Lett ; 589(23): 3631-9, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26497085

RESUMEN

Our understanding of how metabolic switches occur in the failing heart is still limited. Here, we report the emblematic pattern of metabolic alternations in two different mouse models. PP2Acα deficient hearts exhibited a dramatic decrease in the levels of mRNA encoding for transporters and enzymes involved in glucose utilization, which compensated by higher expression levels of genes controlling fatty acid utilization. These features were partly reproduced in cultured PP2Acα KD cardiomyocytes. Equivalently, a decrease in the expression of most of the transporters and enzymes controlling both glucose and fatty acid metabolism were observed in TAC model.


Asunto(s)
Corazón/fisiopatología , Miocardio/metabolismo , Presión/efectos adversos , Proteína Fosfatasa 2/deficiencia , Adaptación Fisiológica , Animales , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Metabolismo Energético , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patología , Hipertrofia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Especificidad de Órganos , Estrés Fisiológico , Factores de Tiempo
16.
Biomolecules ; 5(3): 1284-301, 2015 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-26131975

RESUMEN

Protein phosphatase 2A (PP2A) is one of the most abundant intracellular serine/threonine (Ser/Thr) phosphatases accounting for 1% of the total cellular protein content. PP2A is comprised of a heterodimeric core enzyme and a substrate-specific regulatory subunit. Potentially, at least seventy different compositions of PP2A exist because of variable regulatory subunit binding that accounts for various activity modulating numerous cell functions. Due to the constitutive phosphatase activity present inside cells, a sensitive assay is required to detect the changes of PP2A activity under various experimental conditions. We optimized a fluorescence assay (DIFMU assay) by combining it with prior anti-PP2A immunoprecipitation to quantify PP2A-specific phosphatase activity. It is also known that prior exposure to lipopolysaccharides (LPS) induces "immune tolerance" of the cells to subsequent stimulation. Herein we report that PP2A activity is upregulated in tolerized peritoneal macrophages, corresponding to decreased TNF-α secretion upon second LPS stimulation. We further examined the role of PP2A in the tolerance effect by using PP2ACαl°xl°x;lyM-Cre conditional knockout macrophages. We found that PP2A phosphatase activity cannot be further increased by tolerance. TNF-α secretion from tolerized PP2ACαl°xl°x;lyM-Cre macrophages is higher than tolerized control macrophages. Furthermore, we showed that the increased TNF-α secretion may be due to an epigenetic transcriptionally active signature on the promoter of TNF-α gene rather than regulation of the NFκB/IκB signaling pathway. These results suggest a role for increased PP2A activity in the regulation of immune tolerance.


Asunto(s)
Pruebas de Enzimas/métodos , Tolerancia Inmunológica/efectos de los fármacos , Inmunoprecipitación , Lipopolisacáridos/farmacología , Proteína Fosfatasa 2/metabolismo , Animales , Cromatina/metabolismo , Epigénesis Genética/efectos de los fármacos , Técnicas de Inactivación de Genes , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Transducción de Señal/efectos de los fármacos , Espectrometría de Fluorescencia , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
17.
Am J Respir Crit Care Med ; 190(11): 1229-42, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25341065

RESUMEN

RATIONALE: α1-Antitrypsin (A1AT) was identified as a plasma protease inhibitor; however, it is now recognized as a multifunctional protein that modulates immunity, inflammation, proteostasis, apoptosis, and cellular senescence. Like A1AT, protein phosphatase 2A (PP2A), a major serine-threonine phosphatase, regulates similar biologic processes and plays a key role in chronic obstructive pulmonary disease. OBJECTIVES: Given their common effects, this study investigated whether A1AT acts via PP2A to alter tumor necrosis factor (TNF) signaling, inflammation, and proteolytic responses in this disease. METHODS: PP2A activity was measured in peripheral blood neutrophils from A1AT-deficient (PiZZ) and healthy (PiMM) individuals and in alveolar macrophages from normal (60 mg/kg) and high-dose (120 mg/kg) A1AT-treated PiZZ subjects. PP2A activation was assessed in human neutrophils, airway epithelial cells, and peripheral blood monocytes treated with plasma purified A1AT protein. Similarly, lung PP2A activity was measured in mice administered intranasal A1AT. PP2A was silenced in lung epithelial cells treated with A1AT and matrix metalloproteinase and cytokine production was then measured following TNF-α stimulation. MEASUREMENTS AND MAIN RESULTS: PP2A was significantly lower in neutrophils isolated from PiZZ compared with PiMM subjects. A1AT protein activated PP2A in human alveolar macrophages, monocytes, neutrophils, airway epithelial cells, and in mouse lungs. This activation required functionally active A1AT protein and protein tyrosine phosphatase 1B expression. A1AT treatment acted via PP2A to prevent p38 and IκBα phosphorylation and matrix metalloproteinase and cytokine induction in TNF-α-stimulated epithelial cells. CONCLUSIONS: Together, these data indicate that A1AT modulates PP2A to counter inflammatory and proteolytic responses induced by TNF signaling in the lung.


Asunto(s)
Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Proteína Fosfatasa 2/efectos de los fármacos , Inhibidores de Serina Proteinasa/farmacología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , alfa 1-Antitripsina/farmacología , Adulto , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/inmunología , Pulmón/inmunología , Masculino , Ratones , Persona de Mediana Edad , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Inhibidores de Serina Proteinasa/deficiencia , Inhibidores de Serina Proteinasa/metabolismo , Fumar/fisiopatología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , alfa 1-Antitripsina/metabolismo
18.
Nature ; 506(7486): 52-7, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24476824

RESUMEN

Recent clinical trials showed that targeting of inhibitory receptors on T cells induces durable responses in a subset of cancer patients, despite advanced disease. However, the regulatory switches controlling T-cell function in immunosuppressive tumours are not well understood. Here we show that such inhibitory mechanisms can be systematically discovered in the tumour microenvironment. We devised an in vivo pooled short hairpin RNA (shRNA) screen in which shRNAs targeting negative regulators became highly enriched in murine tumours by releasing a block on T-cell proliferation upon tumour antigen recognition. Such shRNAs were identified by deep sequencing of the shRNA cassette from T cells infiltrating tumour or control tissues. One of the target genes was Ppp2r2d, a regulatory subunit of the PP2A phosphatase family. In tumours, Ppp2r2d knockdown inhibited T-cell apoptosis and enhanced T-cell proliferation as well as cytokine production. Key regulators of immune function can therefore be discovered in relevant tissue microenvironments.


Asunto(s)
Inmunoterapia , Terapia Molecular Dirigida , Proteína Fosfatasa 2/metabolismo , Microambiente Tumoral/inmunología , Animales , Antígenos de Neoplasias/inmunología , Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados
19.
Cancer Lett ; 345(1): 124-31, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24333728

RESUMEN

P-glycoprotein (P-gp)/ABCB1 is a key molecule of multidrug resistance in cancer. Protein phosphatase (PP) 2A, regulatory subunit B, gamma (PPP2R3C), which is a regulatory subunit of PP2A and PP5, was identified as a binding candidate to P-gp. Immunoprecipitation-western blotting revealed that PP5 and PPP2R3C were coprecipitated with P-gp, while PP2A was not. PP5/PPP2R3C dephosphorylated protein kinase A/protein kinase C-phosphorylation of P-gp. Knockdown of PP5 and/or PPP2R3C increased P-gp expression and lowered the sensitivity to vincristine and doxorubicin. Consequently, our results indicate that PP5/PPP2R3C negatively regulates P-gp expression and function.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Glicoproteínas/metabolismo , Proteína Fosfatasa 2/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Femenino , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Técnicas de Silenciamiento del Gen , Glicoproteínas/deficiencia , Glicoproteínas/genética , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosforilación , Proteína Quinasa C/metabolismo , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Transfección , Vincristina/farmacología
20.
Biosci Biotechnol Biochem ; 77(9): 1814-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24018669

RESUMEN

This study shows that the ECM degradation-associated pathway, including uPA and tPA and the downstream MMP-2/-9 protein, was significantly suppressed in HA22T cells treated with a Zanthoxylum avicennae extract (YBBE). The endogenous inhibitors, including TIMP-1/-2 and PAI-1, were enhanced in HA22T cells by the YBBE treatment. The expression of MMP-2/-9 and TIMP-1/-2 was respectively assessed by using RT-PCR and a zymography assay. The mRNA levels and enzymatic activity of MMP-2/-9 were down-regulated by the YBBE treatment in a dose-dependent manner, while the TIMP-1/-2 levels were conversely markedly increased. The PP2A siRNA or PP2A inhibitor totally reversed the YBBE effects, confirming the essential role of PP2A in YBBE inhibiting the HA22T cell migration and invasion effects. Xenografted animal experiments on nude mice demonstrated similiar results to the in vitro system. Both the in vitro and in vivo models clearly demonstrate that YBBE inhibited the highly metastatic HA22T liver cancer cell migration and invasion effects through PP2A activation.


Asunto(s)
Carcinoma Hepatocelular/patología , Movimiento Celular/efectos de los fármacos , Metaloproteinasas de la Matriz/metabolismo , Extractos Vegetales/farmacología , Activadores Plasminogénicos/metabolismo , Proteína Fosfatasa 2/metabolismo , Zanthoxylum/química , Animales , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Neoplasias Hepáticas/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Ácido Ocadaico/farmacología , Proteína Fosfatasa 2/deficiencia , Proteína Fosfatasa 2/genética , Inhibidores Tisulares de Metaloproteinasas/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...