Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
J Biol Chem ; 300(5): 107268, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582449

RESUMEN

Dysregulation of phosphorylation-dependent signaling is a hallmark of tumorigenesis. Protein phosphatase 2 (PP2A) is an essential regulator of cell growth. One scaffold subunit (A) binds to a catalytic subunit (C) to form a core AC heterodimer, which together with one of many regulatory (B) subunits forms the active trimeric enzyme. The combinatorial number of distinct PP2A complexes is large, which results in diverse substrate specificity and subcellular localization. The detailed mechanism of PP2A assembly and regulation remains elusive and reports about an important role of methylation of the carboxy terminus of PP2A C are conflicting. A better understanding of the molecular underpinnings of PP2A assembly and regulation is critical to dissecting PP2A function in physiology and disease. Here, we combined biochemical reconstitution, mass spectrometry, X-ray crystallography, and functional assays to characterize the assembly of trimeric PP2A. In vitro studies demonstrated that methylation of the carboxy-terminus of PP2A C was dispensable for PP2A assembly in vitro. To corroborate these findings, we determined the X-ray crystal structure of the unmethylated PP2A Aα-B56ε-Cα trimer complex to 3.1 Å resolution. The experimental structure superimposed well with an Alphafold2Multimer prediction of the PP2A trimer. We then predicted models of all canonical PP2A complexes providing a framework for structural analysis of PP2A. In conclusion, methylation was dispensable for trimeric PP2A assembly and integrative structural biology studies of PP2A offered predictive models for all canonical PP2A complexes.


Asunto(s)
Proteína Fosfatasa 2 , Humanos , Dominio Catalítico , Cristalografía por Rayos X , Metilación , Multimerización de Proteína , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/química
2.
J Chem Phys ; 158(21)2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37260014

RESUMEN

Mutations in protein phosphatase 2A (PP2A) are connected to intellectual disability and cancer. It has been hypothesized that these mutations might disrupt the autoinhibition and phosphorylation-induced activation of PP2A. Since they are located far from both the active and substrate binding sites, it is unclear how they exert their effect. We performed allosteric pathway analysis based on molecular dynamics simulations and combined it with biochemical experiments to investigate the autoinhibition of PP2A. In the wild type (WT), the C-arm of the regulatory subunit B56δ obstructs the active and substrate binding sites exerting a dual autoinhibition effect. We find that the disease mutant, E198K, severely weakens the allosteric pathways that stabilize the C-arm in the WT. Instead, the strongest allosteric pathways in E198K take a different route that promotes exposure of the substrate binding site. To facilitate the allosteric pathway analysis, we introduce a path clustering algorithm for lumping pathways into channels. We reveal remarkable similarities between the allosteric channels of E198K and those in phosphorylation-activated WT, suggesting that the autoinhibition can be alleviated through a conserved mechanism. In contrast, we find that another disease mutant, E200K, which is in spatial proximity of E198, does not repartition the allosteric pathways leading to the substrate binding site; however, it may still induce exposure of the active site. This finding agrees with our biochemical data, allowing us to predict the activity of PP2A with the phosphorylated B56δ and provide insight into how disease mutations in spatial proximity alter the enzymatic activity in surprisingly different mechanisms.


Asunto(s)
Proteína Fosfatasa 2 , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo , Fosforilación/genética , Dominios Proteicos , Mutación , Unión Proteica
3.
New Phytol ; 236(5): 1762-1778, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36073540

RESUMEN

The various combinations and regulations of different subunits of phosphatase PP2A holoenzymes underlie their functional complexity and importance. However, molecular mechanisms governing the assembly of PP2A complex in response to external or internal signals remain largely unknown, especially in Arabidopsis thaliana. We found that the phosphorylation status of Bß of PP2A acts as a switch to regulate the activity of PP2A. In the absence of ethylene, phosphorylated Bß leads to an inactivation of PP2A; the substrate EIR1 remains to be phosphorylated, preventing the EIR1-mediated auxin transport in epidermis, leading to normal root growth. Upon ethylene treatment, the dephosphorylated Bß mediates the formation of the A2-C4-Bß protein complex to activate PP2A, resulting in the dephosphorylation of EIR1 to promote auxin transport in epidermis of elongation zone, leading to root growth inhibition. Altogether, our research revealed a novel molecular mechanism by which the dephosphorylation of Bß subunit switches on PP2A activity to dephosphorylate EIR1 to establish EIR1-mediated auxin transport in the epidermis in elongation zone for root growth inhibition in response to ethylene.


Asunto(s)
Arabidopsis , Monoéster Fosfórico Hidrolasas , Fosforilación , Monoéster Fosfórico Hidrolasas/metabolismo , Etilenos/metabolismo , Arabidopsis/metabolismo , Ácidos Indolacéticos/farmacología , Ácidos Indolacéticos/metabolismo , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo
4.
J Biol Chem ; 297(1): 100908, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34171357

RESUMEN

The cAMP response element-binding protein (CREB) is an important regulator of cell growth, metabolism, and synaptic plasticity. CREB is activated through phosphorylation of an evolutionarily conserved Ser residue (S133) within its intrinsically disordered kinase-inducible domain (KID). Phosphorylation of S133 in response to cAMP, Ca2+, and other stimuli triggers an association of the KID with the KID-interacting (KIX) domain of the CREB-binding protein (CBP), a histone acetyl transferase (HAT) that promotes transcriptional activation. Here we addressed the mechanisms of CREB attenuation following bursts in CREB phosphorylation. We show that phosphorylation of S133 is reversed by protein phosphatase 2A (PP2A), which is recruited to CREB through its B56 regulatory subunits. We found that a B56-binding site located at the carboxyl-terminal boundary of the KID (BS2) mediates high-affinity B56 binding, while a second binding site (BS1) located near the amino terminus of the KID mediates low affinity binding enhanced by phosphorylation of adjacent casein kinase (CK) phosphosites. Mutations that diminished B56 binding to BS2 elevated both basal and stimulus-induced phosphorylation of S133, increased CBP interaction with CREB, and potentiated the expression of CREB-dependent reporter genes. Cells from mice harboring a homozygous CrebE153D mutation that disrupts BS2 exhibited increased S133 phosphorylation stoichiometry and elevated transcriptional bursts to cAMP. These findings provide insights into substrate targeting by PP2A holoenzymes and establish a new mechanism of CREB attenuation that has implications for understanding CREB signaling in cell growth, metabolism, synaptic plasticity, and other physiologic contexts.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteína Fosfatasa 2/química , Animales , Sitios de Unión , Células Cultivadas , Células HeLa , Humanos , Ratones , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/metabolismo , Transducción de Señal , Activación Transcripcional
5.
Clin Sci (Lond) ; 135(13): 1545-1556, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34192314

RESUMEN

Tumor initiation is driven by oncogenes that activate signaling networks for cell proliferation and survival involving protein phosphorylation. Protein kinases in these pathways have proven to be effective targets for pharmaceutical inhibitors that have progressed to the clinic to treat various cancers. Here, we offer a narrative about the development of small molecule modulators of the protein Ser/Thr phosphatase 2A (PP2A) to reduce the activation of cell proliferation and survival pathways. These novel drugs promote the assembly of select heterotrimeric forms of PP2A that act to limit cell proliferation. We discuss the potential for the near-term translation of this approach to the clinic for cancer and other human diseases.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Proteína Fosfatasa 2/antagonistas & inhibidores , Regulación Alostérica , Animales , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/patología , Fosforilación , Conformación Proteica , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo , Transducción de Señal , Relación Estructura-Actividad
6.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836590

RESUMEN

Reactive oxygen species (ROS) can cause cellular damage and promote cancer development. Besides such harmful consequences of overproduction of ROS, all cells utilize ROS for signaling purposes and stabilization of cell homeostasis. In particular, the latter is supported by the NADPH oxidase 4 (Nox4) that constitutively produces low amounts of H2O2 By that mechanism, Nox4 forces differentiation of cells and prevents inflammation. We hypothesize a constitutive low level of H2O2 maintains basal activity of cellular surveillance systems and is unlikely to be cancerogenic. Utilizing two different murine models of cancerogen-induced solid tumors, we found that deletion of Nox4 promotes tumor formation and lowers recognition of DNA damage. Nox4 supports phosphorylation of H2AX (γH2AX), a prerequisite of DNA damage recognition, by retaining a sufficiently low abundance of the phosphatase PP2A in the nucleus. The underlying mechanism is continuous oxidation of AKT by Nox4. Interaction of oxidized AKT and PP2A captures the phosphatase in the cytosol. Absence of Nox4 facilitates nuclear PP2A translocation and dephosphorylation of γH2AX. Simultaneously AKT is left phosphorylated. Thus, in the absence of Nox4, DNA damage is not recognized and the increased activity of AKT supports proliferation. The combination of both events results in genomic instability and promotes tumor formation. By identifying Nox4 as a protective source of ROS in cancerogen-induced cancer, we provide a piece of knowledge for understanding the role of moderate production of ROS in preventing the initiation of malignancies.


Asunto(s)
Carcinógenos/toxicidad , NADPH Oxidasa 4/genética , Neoplasias/inducido químicamente , Animales , Núcleo Celular/metabolismo , Citosol/metabolismo , Daño del ADN , Inestabilidad Genómica , Ratones , NADPH Oxidasa 4/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Oxidación-Reducción , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo , Subunidades de Proteína , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal
7.
Biomolecules ; 10(11)2020 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-33266510

RESUMEN

Protein phosphorylation is a post-translational modification essential for the control of the activity of most enzymes in the cell. This protein modification results from a fine-tuned balance between kinases and phosphatases. PP2A is one of the major serine/threonine phosphatases that is involved in the control of a myriad of different signaling cascades. This enzyme, often misregulated in cancer, is considered a tumor suppressor. In this review, we will focus on PP2A-B55, a particular holoenzyme of the family of the PP2A phosphatases whose specific role in cancer development and progression has only recently been highlighted. The discovery of the Greatwall (Gwl)/Arpp19-ENSA cascade, a new pathway specifically controlling PP2A-B55 activity, has been shown to be frequently altered in cancer. Herein, we will review the current knowledge about the mechanisms controlling the formation and the regulation of the activity of this phosphatase and its misregulation in cancer.


Asunto(s)
Neoplasias/enzimología , Neoplasias/genética , Proteína Fosfatasa 2/farmacocinética , Animales , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
8.
Biochem Soc Trans ; 48(5): 2015-2027, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33125487

RESUMEN

Phosphoprotein Phosphatases (PPPs) are enzymes highly conserved from yeast and human and catalyze the majority of the serine and threonine dephosphorylation in cells. To achieve substrate specificity and selectivity, PPPs form multimeric holoenzymes consisting of catalytic, structural/scaffolding, and regulatory subunits. For the Protein Phosphatase 2A (PP2A)-subfamily of PPPs, holoenzyme assembly is at least in part regulated by an unusual carboxyl-terminal methyl-esterification, commonly referred to as 'methylation'. Carboxyl-terminal methylation is catalyzed by Leucine carboxyl methyltransferase-1 (LCMT1) that utilizes S-adenosyl-methionine (SAM) as the methyl donor and removed by protein phosphatase methylesterase 1 (PME1). For PP2A, methylation dictates regulatory subunit selection and thereby downstream phosphorylation signaling. Intriguingly, there are four families of PP2A regulatory subunits, each exhibiting different levels of methylation sensitivity. Thus, changes in PP2A methylation stoichiometry alters the complement of PP2A holoenzymes in cells and creates distinct modes of kinase opposition. Importantly, selective inactivation of PP2A signaling through the deregulation of methylation is observed in several diseases, most prominently Alzheimer's disease (AD). In this review, we focus on how carboxyl-terminal methylation of the PP2A subfamily (PP2A, PP4, and PP6) regulates holoenzyme function and thereby phosphorylation signaling, with an emphasis on AD.


Asunto(s)
Enzimas/química , Regulación de la Expresión Génica , Fosfoproteínas/química , Proteína Fosfatasa 2/química , Enfermedad de Alzheimer/metabolismo , Animales , Catálisis , Dominio Catalítico , Dimerización , Holoenzimas/química , Humanos , Metilación , Ratones , Mutación , Fosforilación , Conformación Proteica , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Especificidad por Sustrato
9.
Cell ; 181(3): 688-701.e16, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32315618

RESUMEN

Impairment of protein phosphatases, including the family of serine/threonine phosphatases designated PP2A, is essential for the pathogenesis of many diseases, including cancer. The ability of PP2A to dephosphorylate hundreds of proteins is regulated by over 40 specificity-determining regulatory "B" subunits that compete for assembly and activation of heterogeneous PP2A heterotrimers. Here, we reveal how a small molecule, DT-061, specifically stabilizes the B56α-PP2A holoenzyme in a fully assembled, active state to dephosphorylate selective substrates, such as its well-known oncogenic target, c-Myc. Our 3.6 Å structure identifies molecular interactions between DT-061 and all three PP2A subunits that prevent dissociation of the active enzyme and highlight inherent mechanisms of PP2A complex assembly. Thus, our findings provide fundamental insights into PP2A complex assembly and regulation, identify a unique interfacial stabilizing mode of action for therapeutic targeting, and aid in the development of phosphatase-based therapeutics tailored against disease specific phospho-protein targets.


Asunto(s)
Proteína Fosfatasa 2/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Activadores de Enzimas/metabolismo , Células HEK293 , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Proteína Fosfatasa 2/química , Subunidades de Proteína
10.
Cell Rep ; 30(9): 3164-3170.e3, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130915

RESUMEN

Aberrant hyperphosphorylation of the protein phosphatase 2A catalytic subunit (PP2Ac) at Tyr307 has been associated with aggressive disease and poor clinical outcome in multiple cancers. However, the study of reversible phosphorylation at this site has relied entirely upon the use of antibodies-most prominently, the clone E155. Here, we provide evidence that the E155 and F-8 phospho-Tyr307 antibodies cannot differentiate between phosphorylated and unphosphorylated forms of PP2Ac. The form of PP2Ac bound by these antibodies in H358 cells is unphosphorylated at the C-terminal tail. Furthermore, these antibodies are sensitive to additional protein modifications that occur near Tyr307, including Thr304 phosphorylation and Leu309 methylation, when these post-translational modifications are present. Thus, studies that used these antibodies to report PP2Ac hyperphosphorylation require reinterpretation, as these antibodies cannot be reliably used as readouts for a single PP2Ac post-translational modification (PTM) change.


Asunto(s)
Anticuerpos/metabolismo , Fosfotirosina/metabolismo , Proteína Fosfatasa 2/metabolismo , Investigación , Fosfatasa Alcalina/metabolismo , Secuencia de Aminoácidos , Línea Celular , Línea Celular Tumoral , Humanos , Leucina/metabolismo , Metilación , Mutación/genética , Péptidos/química , Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/genética , Procesamiento Proteico-Postraduccional , Vanadatos/farmacología
11.
Adv Cancer Res ; 144: 55-93, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31349904

RESUMEN

PP2A is a highly conserved eukaryotic serine/threonine protein phosphatase of the PPP family of phosphatases with fundamental cellular functions. In cells, PP2A targets specific subcellular locations and substrates by forming heterotrimeric holoenzymes, where a core dimer consisting of scaffold (A) and catalytic (C) subunits complexes with one of many B regulatory subunits. PP2A plays a key role in positively and negatively regulating a myriad of cellular processes, as it targets a very sizable fraction of the cellular substrates phosphorylated on Ser/Thr residues. This review focuses on insights made toward the understanding on how the subunit composition and structure of PP2A holoenzymes mediates substrate specificity, the role of substrate modulation in the signaling of cellular division, growth, and differentiation, and its deregulation in cancer.


Asunto(s)
Neoplasias/enzimología , Neoplasias/patología , Proteína Fosfatasa 2/metabolismo , Animales , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Holoenzimas/química , Holoenzimas/metabolismo , Humanos , Fosforilación , Proteína Fosfatasa 2/química , Transducción de Señal , Especificidad por Sustrato
12.
Mol Cancer Ther ; 18(3): 556-566, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30679389

RESUMEN

LB-100 is an experimental cancer therapeutic with cytotoxic activity against cancer cells in culture and antitumor activity in animals. The first phase I trial (NCT01837667) evaluating LB-100 recently concluded that safety and efficacy parameters are favorable for further clinical testing. Although LB-100 is widely reported as a specific inhibitor of serine/threonine phosphatase 2A (PP2AC/PPP2CA:PPP2CB), we could find no experimental evidence in the published literature demonstrating the specific engagement of LB-100 with PP2A in vitro, in cultured cells, or in animals. Rather, the premise for LB-100 targeting PP2AC is derived from studies that measure phosphate released from a phosphopeptide (K-R-pT-I-R-R) or inferred from the ability of LB-100 to mimic activity previously reported to result from the inhibition of PP2AC by other means. PP2AC and PPP5C share a common catalytic mechanism. Here, we demonstrate that the phosphopeptide used to ascribe LB-100 specificity for PP2A is also a substrate for PPP5C. Inhibition assays using purified enzymes demonstrate that LB-100 is a catalytic inhibitor of both PP2AC and PPP5C. The structure of PPP5C cocrystallized with LB-100 was solved to a resolution of 1.65Å, revealing that the 7-oxabicyclo[2.2.1]heptane-2,3-dicarbonyl moiety coordinates with the metal ions and key residues that are conserved in both PP2AC and PPP5C. Cell-based studies revealed some known actions of LB-100 are mimicked by the genetic disruption of PPP5C These data demonstrate that LB-100 is a catalytic inhibitor of both PP2AC and PPP5C and suggest that the observed antitumor activity might be due to an additive effect achieved by suppressing both PP2A and PPP5C.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/química , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/química , Fosfoproteínas Fosfatasas/química , Piperazinas/química , Proteína Fosfatasa 2/química , Secuencia de Aminoácidos/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Catálisis , Dominio Catalítico/efectos de los fármacos , Línea Celular Tumoral , Humanos , Metales/química , Metilación , Mutagénesis Sitio-Dirigida , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Piperazinas/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/genética
13.
Cancer Res ; 79(1): 209-219, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389701

RESUMEN

In cancer, kinases are often activated and phosphatases suppressed, leading to aberrant activation of signaling pathways driving cellular proliferation, survival, and therapeutic resistance. Although pancreatic ductal adenocarcinoma (PDA) has historically been refractory to kinase inhibition, therapeutic activation of phosphatases is emerging as a promising strategy to restore balance to these hyperactive signaling cascades. In this study, we hypothesized that phosphatase activation combined with kinase inhibition could deplete oncogenic survival signals to reduce tumor growth. We screened PDA cell lines for kinase inhibitors that could synergize with activation of protein phosphatase 2A (PP2A), a tumor suppressor phosphatase, and determined that activation of PP2A and inhibition of mTOR synergistically increase apoptosis and reduce oncogenic phenotypes in vitro and in vivo. This combination treatment resulted in suppression of AKT/mTOR signaling coupled with reduced expression of c-MYC, an oncoprotein implicated in tumor progression and therapeutic resistance. Forced expression of c-MYC or loss of PP2A B56α, the specific PP2A subunit shown to negatively regulate c-MYC, increased resistance to mTOR inhibition. Conversely, decreased c-MYC expression increased the sensitivity of PDA cells to mTOR inhibition. Together, these studies demonstrate that combined targeting of PP2A and mTOR suppresses proliferative signaling and induces cell death and implicates this combination as a promising therapeutic strategy for patients with PDA. SIGNIFICANCE: These findings present a combinatorial strategy targeting serine/threonine protein phosphatase PP2A and mTOR in PDA, a cancer for which there are currently no targeted therapeutic options.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/1/209/F1.large.jpg.


Asunto(s)
Benzoxazoles/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Sinergismo Farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Apoptosis , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Activación Enzimática , Humanos , Ratones , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteína Fosfatasa 2/química , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-myc/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(7): 595-599, 2018 Jul.
Artículo en Chino | MEDLINE | ID: mdl-30381122

RESUMEN

Objective To investigate the role of demethylation of protein phosphatase 2A catalytic subunit (PP2Ac) on M1 macrophage polarization. Methods THP-1 cells were induced to differentiate into M0 macrophages with phorbol ester (PMA) for 24 hours, and then stimulated to differentiate into M1 macrophages induced by lipopolysaccharide (LPS) and interferon-γ (IFN-γ) for 48 hours. The administration group and the solvent control group were respectively cultured with 0.5 µmol/L ABL127 and the same volume of DMSO for 48 hours on M1 macrophage polarization models. Inverted microscope was used to observe the morphological changes of macrophages. The mRNA expression levels of cyclooxygenase 2 (COX-2), tumor necrosis factor α (TNF-α), interleukin 6 (IL-6) and C-X-C motif chemokine ligand 10 (CXCL10) in M1 macrophages were detected by quantitative real-time PCR. M1 macrophage phagocytosis was determined by neutral red uptake assay. Western blot analysis was applied to test the level of PP2Ac demethylation. Results In the differentiation of M0 into M1 macrophages, the cells were elongated and became spindle-shaped. Furthermore, the expression of M1 macrophage markers including COX-2, TNF-α, IL-6 and CXCL10 significantly increased and phagocytosis was enhanced, but PP2Ac demethylation was decreased. Compared with the DMSO group, PP2Ac demethylation decreased while spindle cells, phagocytosis, and the mRNA levels of COX-2, TNF-α, IL-6 and CXCL10 significantly increased in the administration group with 0.5 µmol/L ABL127. Conclusion The inhibition of PP2Ac demethylation promotes the differentiation of M1 macrophages, and increases the expression of pro-inflammatory cytokines and the phagocytosis.


Asunto(s)
Polaridad Celular , Desmetilación , Macrófagos/citología , Proteína Fosfatasa 2/química , Dominio Catalítico , Quimiocina CXCL10/metabolismo , Ciclooxigenasa 2/metabolismo , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos , Macrófagos/enzimología , Células THP-1 , Factor de Necrosis Tumoral alfa/metabolismo
15.
Diabetes ; 67(8): 1524-1537, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29764860

RESUMEN

Women gain weight and their diabetes risk increases as they transition through menopause; these changes can be partly reversed by hormone therapy. However, the underlying molecular mechanisms mediating these effects are unknown. A novel knock-in mouse line with the selective blockade of the membrane-initiated estrogen receptor (ER) pathway was used, and we found that the lack of this pathway precipitated excessive weight gain and glucose intolerance independent of food intake and that this was accompanied by impaired adaptive thermogenesis and reduced physical activity. Notably, the central activation of protein phosphatase (PP) 2A improved metabolic disorders induced by the lack of membrane-initiated ER signaling. Furthermore, the antiobesity effect of estrogen replacement in a murine menopause model was abolished by central PP2A inactivation. These findings define a critical role for membrane-initiated ER signaling in metabolic homeostasis via the central action of PP2A.


Asunto(s)
Receptor alfa de Estrógeno/agonistas , Terapia de Reemplazo de Estrógeno , Intolerancia a la Glucosa/prevención & control , Menopausia , Obesidad/prevención & control , Proteína Fosfatasa 2/metabolismo , Transducción de Señal/efectos de los fármacos , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Adiposidad/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Activación Enzimática/efectos de los fármacos , Estradiol/farmacología , Estradiol/uso terapéutico , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Técnicas de Sustitución del Gen , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Ovariectomía , Mutación Puntual , Proteína Fosfatasa 2/química
16.
World J Surg ; 42(11): 3771-3778, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29796729

RESUMEN

BACKGROUND: The functional loss of the tumor suppressor protein phosphatase 2A (PP2A) occurs in a wide variety of human cancers including colorectal cancer (CRC), and SET overexpression has been reported as a key contributing mechanism to inhibit PP2A. Although SET binding protein 1 (SETBP1) overexpression and gain of function mutations have been described in several hematological malignancies as common events that increase the expression levels of the PP2A inhibitor SET, thereby leading to PP2A inactivation, the potential existence of SETBP1 alterations in CRC still remains unexplored. METHODS: We studied the expression profile of SETBP1 by Western blot in a set of CRC cell lines and patient samples. Moreover, we performed co-immunoprecipitation assays to analyze the formation of the previously reported SETBP1-SET-PP2A inhibitory complex. Furthermore, we evaluated the mutational status of SETBP1 by pyrosequencing assays in a cohort of 55 CRC patients with metastatic disease after the immunohistochemical characterization of SET and p-PP2A expression in this cohort. RESULTS: We found high SETBP1 expression in several CRC lines but only in two of the patients analyzed. In addition, we demonstrated the formation of the SETBP1-SET-PP2A heterotrimeric complex in CRC cells. However, we failed to detect SETBP1 mutations in any of the CRC patient samples included in the study. CONCLUSIONS: Our results suggest that SETBP1 expression is mainly similar o lower in colorectal cancer tissue compared to normal colonic mucosa. However, its overexpression is a low prevalent alteration which could contribute to inhibit PP2A in CRC through the formation of a SETBP1-SET-PP2A complex in some CRC patients. Moreover, SETBP1 mutations are, if exist, rare events in CRC patients.


Asunto(s)
Proteínas Portadoras/genética , Neoplasias Colorrectales/genética , Mutación , Proteínas Nucleares/genética , Proteína Fosfatasa 2/antagonistas & inhibidores , Adulto , Anciano , Proteínas Portadoras/química , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas de Unión al ADN , Femenino , Chaperonas de Histonas/química , Chaperonas de Histonas/fisiología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Nucleares/química , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/fisiología , Factores de Transcripción/química , Factores de Transcripción/fisiología
17.
Med Hypotheses ; 113: 9-12, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29523304

RESUMEN

Small viral proteins with cationic domains can be involved in multiple biological processes including cell penetration or interaction with intracellular targets. Within the last two decades several reports indicated that the C-terminus of HIV-1 Vpr is a cell penetrating sequence, a PP2A-dependent death domain and also displays toxicity against Gram-negative E. coli. Interestingly, HIV-1 Vpr, as well as some cationic proteins encoded by different viruses, share similar physical properties with the unique anti-microbial human cathelicidin LL37 peptide. Consistent with these observations, the Viral Quinta Columna Hypothesis predicts that virally-encoded cationic peptides encoded by multiple viruses may at the same time i) behave as new cathelicidin-like viral positive effectors of innate immunity, mainly through electrostatic interactions with microbial walls, and also display specific toxic cellular effects through interactions with specific intracellular targets such as PP2A proteins. In this context, virally-encoded cationic peptides, potentially detectable in biological fluids, may define a new paradigm for a viral control of homeostasis. Finally, we can also predict that characterization of virally encoded sequences with anti-infective effects may serve as template for the design of new efficient therapeutics polypeptides.


Asunto(s)
Infecciones por Escherichia coli/diagnóstico , Infecciones por VIH/diagnóstico , VIH-1 , Péptidos Catiónicos Antimicrobianos/química , Cationes , Infecciones por Escherichia coli/microbiología , Infecciones por VIH/virología , Homeostasis , Humanos , Infectología , Modelos Teóricos , Péptidos/química , Dominios Proteicos , Proteína Fosfatasa 2/química , Electricidad Estática , Catelicidinas
18.
J Mol Cell Cardiol ; 115: 20-31, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29294329

RESUMEN

BACKGROUND: Type 2A protein phosphatase (PP2A) enzymes are serine/threonine phosphatases which comprise a scaffold A subunit, a regulatory B subunit and a catalytic C subunit, and have been implicated in the dephosphorylation of multiple cardiac phosphoproteins. B subunits determine subcellular targeting, substrate specificity and catalytic activity, and can themselves be regulated by post-translational modifications. We explored potential ß-adrenergic regulation of PP2A in cardiomyocytes through phosphorylation of the regulatory B subunit isoform B56δ. METHODS AND RESULTS: Phosphate affinity SDS-PAGE and immunoblot analysis revealed increased phosphorylation of B56δ in adult rat ventricular myocytes (ARVM) exposed to the ß-adrenergic receptor (ßAR) agonist isoprenaline (ISO). Phosphorylation of B56δ occurred at S573, primarily through stimulation of the ß1AR subtype, and was dependent on PKA activity. The functional role of the phosphorylation was explored in ARVM transduced with adenoviruses expressing wild type (WT) or non-phosphorylatable (S573A) B56δ, fused to GFP at the N-terminus. C subunit expression was increased in ARVM expressing GFP-B56δ-WT or GFP-B56δ-S573A, both of which co-immunoprecipitated with endogenous C and A subunits. PP2A activity in cell lysates was increased in response to ISO in ARVM expressing GFP-B56δ-WT but not GFP-B56δ-S573A. Immunoblot analysis of the phosphoproteome in ARVM expressing GFP-B56δ-WT or GFP-B56δ-S573A with antibodies detecting (i) phospho-serine/threonine residues in distinct kinase substrate motifs or (ii) specific phosphorylated residues of functional importance in selected proteins revealed a comparable phosphorylation profile in the absence or presence of ISO stimulation. CONCLUSIONS: In cardiomyocytes, ßAR stimulation induces PKA-mediated phosphorylation of the PP2A regulatory subunit isoform B56δ at S573, which increases associated PP2A catalytic activity. This is likely to regulate the phosphorylation status of specific B56δ-PP2A substrates, which remain to be identified.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Miocardio/enzimología , Fosfoserina/metabolismo , Proteína Fosfatasa 2/metabolismo , Subunidades de Proteína/metabolismo , Adenoviridae/metabolismo , Secuencia de Aminoácidos , Animales , Cardiomegalia/enzimología , Cardiomegalia/patología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Isoproterenol/farmacología , Masculino , Ratones , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/química , Subunidades de Proteína/química , Ratas Wistar
19.
J Biomol Struct Dyn ; 36(12): 3155-3171, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28920551

RESUMEN

A polyglutamine expansion of the N-terminal region of huntingtin (Htt) causes Huntington's disease, a severe neurodegenerative disorder. Htt huge multidomain structure, the presence of disordered regions, and the lack of sequence homologs of known structure, so far prevented structural studies of Htt, making the study of its structure-function relationships very difficult. In this work, the presence and location of five Htt ordered domains (named from Hunt1 to Hunt5) has been detected and the structure of these domains has been predicted for the first time using a combined threading/ab initio modeling approach. This work has led to the identification of a previously undetected HEAT repeats region in the Hunt3 domain. Furthermore, a putative function has been assigned to four out of the five domains. Hunt1 and Hunt5, displaying structural similarity with the regulatory subunit A of protein phosphatase 2A, are predicted to play a role in regulating the phosphorylation status of cellular proteins. Hunt2 and Hunt3 are predicted to be homologs of two yeast importins and to mediate vescicles transport and protein trafficking. Finally, a comprehensive analysis of the Htt interactome has been carried out and is discussed to provide a global picture of the Htt's structure-function relationships.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Secuencias Repetitivas de Aminoácido/genética , Relación Estructura-Actividad , Simulación por Computador , Humanos , Proteína Huntingtina/química , Enfermedad de Huntington/patología , Péptidos/química , Péptidos/genética , Dominios Proteicos , Mapas de Interacción de Proteínas/genética , Proteína Fosfatasa 2/química
20.
Chemistry ; 24(8): 1890-1897, 2018 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-29193344

RESUMEN

Apoptotic signaling pathways are altered in numerous pathologies such as cancer. In this scenario, caspase-9/PP2Acα interaction constitutes a key target with pharmacological interest to re-establish apoptosis in tumor cells. Very recently, a short peptide (C9h) known to disrupt caspase-9/PP2Acα interaction with subsequent apoptosis induction was described. Here, we prepared two sets of mesoporous silica nanoparticles loaded with safranin O (S2) or with C9h peptide (S4) and functionalized with ϵ-polylysine as capping unit. Aqueous suspensions of both nanoparticles showed negligible cargo release whereas in the presence of pronase, a marked delivery of safranin O or C9h was observed. Confocal microscopy studies carried out with HeLa cells indicated that both materials were internalized and were able to release their entrapped cargos. Besides, a marked decrease in HeLa cell viability (ca. 50 %) was observed when treated with C9h-loaded S4 nanoparticles. Moreover, S4 provides peptide protection from degradation additionally allowing for a dose reduction to observe an apoptotic effect when compared with C9h alone or in combination with a cell-penetrating peptide (i.e., Mut3DPT-C9h). Flow cytometry studies, by means of Annexin V-FITC staining, showed the activation of apoptotic pathways in HeLa as a consequence of S4 internalization, release of C9h peptide and disruption of caspase-9/PP2Acα interaction.


Asunto(s)
Nanopartículas/química , Péptidos/química , Polilisina/química , Dióxido de Silicio/química , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Caspasa 9/química , Caspasa 9/metabolismo , Dicroismo Circular , Portadores de Fármacos/síntesis química , Portadores de Fármacos/química , Células HeLa , Humanos , Microscopía Confocal , Péptidos/toxicidad , Fenazinas/química , Fenazinas/toxicidad , Porosidad , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA