Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 3778, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35773251

RESUMEN

PPM1D encodes a serine/threonine phosphatase that regulates numerous pathways including the DNA damage response and p53. Activating mutations and amplification of PPM1D are found across numerous cancer types. GSK2830371 is a potent and selective allosteric inhibitor of PPM1D, but its mechanism of binding and inhibition of catalytic activity are unknown. Here we use computational, biochemical and functional genetic studies to elucidate the molecular basis of GSK2830371 activity. These data confirm that GSK2830371 binds an allosteric site of PPM1D with high affinity. By further incorporating data from hydrogen deuterium exchange mass spectrometry and sedimentation velocity analytical ultracentrifugation, we demonstrate that PPM1D exists in an equilibrium between two conformations that are defined by the movement of the flap domain, which is required for substrate recognition. A hinge region was identified that is critical for switching between the two conformations and was directly implicated in the high-affinity binding of GSK2830371 to PPM1D. We propose that the two conformations represent active and inactive forms of the protein reflected by the position of the flap, and that binding of GSK2830371 shifts the equilibrium to the inactive form. Finally, we found that C-terminal truncating mutations proximal to residue 400 result in destabilization of the protein via loss of a stabilizing N- and C-terminal interaction, consistent with the observation from human genetic data that nearly all PPM1D mutations in cancer are truncating and occur distal to residue 400. Taken together, our findings elucidate the mechanism by which binding of a small molecule to an allosteric site of PPM1D inhibits its activity and provides insights into the biology of PPM1D.


Asunto(s)
Neoplasias , Proteína Fosfatasa 2C , Sitio Alostérico , Aminopiridinas/farmacología , Dipéptidos/farmacología , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Conformación Proteica , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/química , Proteína Fosfatasa 2C/genética , Proteína Fosfatasa 2C/metabolismo , Serina/genética , Serina/metabolismo , Relación Estructura-Actividad
2.
Cell Chem Biol ; 29(7): 1126-1139.e12, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35320734

RESUMEN

Metal-dependent protein phosphatases (PPMs) have essential roles in a variety of cellular processes, including inflammation, proliferation, differentiation, and stress responses, which are intensively investigated in cancer and metabolic diseases. Targeting PPMs to modulate host immunity in response to pathogens is an ambitious proposition. The feasibility of such a strategy is unproven because development of inhibitors against PPMs is challenging and suffers from poor selectivity. Combining a biomimetic modularization strategy with function-oriented synthesis, we design, synthesize and screen more than 500 pseudo-natural products, resulting in the discovery of a potent, selective, and non-cytotoxic small molecule inhibitor for PPM1A, SMIP-30. Inhibition of PPM1A with SMIP-30 or its genetic ablation (ΔPPM1A) activated autophagy through a mechanism dependent on phosphorylation of p62-SQSTM1, which restricted the intracellular survival of Mycobacterium tuberculosis in macrophages and in the lungs of infected mice. SMIP-30 provides proof of concept that PPMs are druggable and promising targets for the development of host-directed therapies against tuberculosis.


Asunto(s)
Autofagia , Proteína Fosfatasa 2C , Tuberculosis , Animales , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Mycobacterium tuberculosis , Proteína Fosfatasa 2C/antagonistas & inhibidores , Tuberculosis/tratamiento farmacológico
3.
Reprod Biomed Online ; 43(2): 161-171, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34210610

RESUMEN

RESEARCH QUESTION: What role does wild-type p53-induced phosphatase 1 (WIP1) play in the regulation of primordial follicle development? DESIGN: WIP1 expression was detected in the ovaries of mice of different ages by western blotting and immunohistochemical staining. Three-day-old neonatal mouse ovaries were cultured in vitro with or without the WIP1 inhibitor GSK2830371 (10 µM) for 4 days. Ovarian morphology, follicle growth and follicle classification were analysed and the PI3K-AKT-mTOR signal pathway and the WIP1-p53-related mitochondrial apoptosis pathway evaluated. RESULTS: WIP1 expression was downregulated with age. Primordial follicles were significantly decreased in the GSK2830371-treated group, without a significant increase in growing follicles. The ratio of growing follicles to primordial follicles was not significantly different between the control and GSK2830371 groups, and no significant variation was observed in the PI3K-AKT-mTOR signal pathway. The inhibition of WIP1 phosphatase accelerated primordial follicle atresia by activating the p53-BAX-caspase-3 pathway. CONCLUSIONS: These findings reveal that WIP1 participates in regulating primordial follicle development and that inhibiting WIP1 phosphatase leads to massive primordial follicle loss via interaction with the p53-BAX-caspase-3 pathway. This might also provide valuable information for understanding decreased ovarian reserve during ovarian ageing.


Asunto(s)
Aminopiridinas/farmacología , Dipéptidos/farmacología , Folículo Ovárico/efectos de los fármacos , Animales , Animales Recién Nacidos , Recuento de Células , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Folículo Ovárico/citología , Folículo Ovárico/fisiología , Ovario/citología , Ovario/efectos de los fármacos , Embarazo , Proteína Fosfatasa 2C/antagonistas & inhibidores
4.
Biochem Pharmacol ; 184: 114362, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33309518

RESUMEN

Aberrations in DNA damage response genes are recognized mediators of tumorigenesis and resistance to chemo- and radiotherapy. While protein phosphatase magnesium-dependent 1 δ (PPM1D), located on the long arm of chromosome 17 at 17q22-23, is a key regulator of cellular responses to DNA damage, amplification, overexpression, or mutation of this gene is important in a wide range of pathologic processes. In this review, we describe the physiologic function of PPM1D, as well as its role in diverse processes, including fertility, development, stemness, immunity, tumorigenesis, and treatment responsiveness. We highlight both the advances and limitations of current approaches to targeting malignant processes mediated by pathogenic alterations in PPM1D with the goal of providing rationale for continued research and development of clinically viable treatment approaches for PPM1D-associated diseases.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Terapia Molecular Dirigida/métodos , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/fisiología , Animales , Ciclo Celular , Daño del ADN , Femenino , Fertilidad/fisiología , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Masculino , Ratones
5.
Cell Mol Life Sci ; 78(6): 2821-2838, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33067654

RESUMEN

Dramatic cellular reorganization in mitosis critically depends on the timely and temporal phosphorylation of a broad range of proteins, which is mediated by the activation of the mitotic kinases and repression of counteracting phosphatases. The mitosis-to-interphase transition, which is termed mitotic exit, involves the removal of mitotic phosphorylation by protein phosphatases. Although protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) drive this reversal in animal cells, the phosphatase network associated with ordered bulk dephosphorylation in mitotic exit is not fully understood. Here, we describe a new mitotic phosphatase relay in which Wip1/PPM1D phosphatase activity is essential for chromosomal passenger complex (CPC) translocation to the anaphase central spindle after release from the chromosome via PP1-mediated dephosphorylation of histone H3T3. Depletion of endogenous Wip1 and overexpression of the phosphatase-dead mutant disturbed CPC translocation to the central spindle, leading to failure of cytokinesis. While Wip1 was degraded in early mitosis, its levels recovered in anaphase and the protein functioned as a Cdk1-counteracting phosphatase at the anaphase central spindle and midbody. Mechanistically, Wip1 dephosphorylated Thr-59 in inner centromere protein (INCENP), which, subsequently bound to MKLP2 and recruited other components to the central spindle. Furthermore, Wip1 overexpression is associated with the overall survival rate of patients with breast cancer, suggesting that Wip1 not only functions as a weak oncogene in the DNA damage network but also as a tumor suppressor in mitotic exit. Altogether, our findings reveal that sequential dephosphorylation of mitotic phosphatases provides spatiotemporal regulation of mitotic exit to prevent tumor initiation and progression.


Asunto(s)
Cromosomas/metabolismo , Mitosis , Proteína Fosfatasa 2C/metabolismo , Huso Acromático/metabolismo , Anafase , Aurora Quinasa B/metabolismo , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/genética , Daño del ADN , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/genética , Cinesinas/metabolismo , Fosforilación , Unión Proteica , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Survivin/metabolismo
6.
J Cell Mol Med ; 24(22): 13463-13471, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33048454

RESUMEN

Protein phosphatase 1B (PPM1B), a member of metal-dependent protein serine/threonine phosphatase family, is involved in the regulation of several signalling pathways. However, our understanding of its substrate interaction and physiological functions is still largely limited. There is no reported PPM1B inhibitor to date. In this study, we identified HN252, a p-terphenyl derivative, as a potent PPM1B inhibitor (Ki  = 0.52 ± 0.06 µM). HN252 binding to PPM1B displayed remarkable and specific inhibition of PPM1B in both in vitro and ex vivo. With the aid of this small molecular inhibitor, we identified 30 proteins' serine/threonine phosphorylation as potential substrates of PPM1B, 5 of which were demonstrated by immunoprecipitation, including one known (CDK2) and 4 novel ones (AKT1, HSP90B, ß-catenin and BRCA1). Furthermore, GO and KEGG analysis of dramatically phosphorylated proteins by PPM1B inhibition indicated that PPM1B plays roles in the regulation of multiple cellular processes and signalling pathways, such as gene transcription, inflammatory regulation, ageing and tumorigenesis. Our work provides novel insights into further investigation of molecular mechanisms of PPM1B.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Fosfatasa 2C/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Espectrometría de Masas , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Fosforilación , Unión Proteica , Proteína Fosfatasa 2C/química , Proteína Fosfatasa 2C/aislamiento & purificación , Proteínas Recombinantes , Relación Estructura-Actividad
7.
Oncogene ; 39(40): 6313-6326, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32848211

RESUMEN

Cancer can metastasize from early lesions without detectable tumors. Despite extensive studies on metastasis in cancer cells from patients with detectable primary tumors, mechanisms for early metastatic dissemination are poorly understood. Her2 promotes breast cancer early dissemination by inhibiting p38, but the downstream pathway in this process was unknown. Using early lesion breast cancer models, we demonstrate that the effect of p38 suppression by Her2 on early dissemination is mediated by MK2 and heat shock protein 27 (Hsp27). The early disseminating cells in the MMTV-Her2 breast cancer model are Her2highp-p38lowp-MK2lowp-Hsp27low, which also exist in human breast carcinoma tissues. Suppression of p38 and MK2 by Her2 reduces MK2-mediated Hsp27 phosphorylation, and unphosphorylated Hsp27 binds to ß-catenin and enhances its phosphorylation by Src, leading to ß-catenin activation and disseminating phenotypes in early lesion breast cancer cells. Pharmacological inhibition of MK2 promotes, while inhibition of a p38 phosphatase Wip1 suppresses, early dissemination in vivo. These findings identify Her2-mediated suppression of the p38-MK2-Hsp27 pathway as a novel mechanism for cancer early dissemination, and provide a basis for new therapies targeting early metastatic dissemination in Her2+ breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Fosfatasa 2C/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Aminopiridinas/farmacología , Aminopiridinas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Femenino , Proteínas de Choque Térmico/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Chaperonas Moleculares/metabolismo , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/farmacología , Piridinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cancer Res ; 18(7): 968-980, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32229503

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) is an invariably fatal brain tumor occurring predominantly in children. Up to 90% of pediatric DIPGs harbor a somatic heterozygous mutation resulting in the replacement of lysine 27 with methionine (K27M) in genes encoding histone H3.3 (H3F3A, 65%) or H3.1 (HIST1H3B, 25%). Several studies have also identified recurrent truncating mutations in the gene encoding protein phosphatase 1D, PPM1D, in 9%-23% of DIPGs. Here, we sought to investigate the therapeutic potential of targeting PPM1D, alone or in combination with inhibitors targeting specific components of DNA damage response pathways in patient-derived DIPG cell lines. We found that GSK2830371, an allosteric PPM1D inhibitor, suppressed the proliferation of PPM1D-mutant, but not PPM1D wild-type DIPG cells. We further observed that PPM1D inhibition sensitized PPM1D-mutant DIPG cells to PARP inhibitor (PARPi) treatment. Mechanistically, combined PPM1D and PARP inhibition show synergistic effects on suppressing a p53-dependent RAD51 expression and the formation of RAD51 nuclear foci, possibly leading to impaired homologous recombination (HR)-mediated DNA repair in PPM1D-mutant DIPG cells. Collectively, our findings reveal the potential role of the PPM1D-p53 signaling axis in the regulation of HR-mediated DNA repair and provide preclinical evidence demonstrating that combined inhibition of PPM1D and PARP1/2 may be a promising therapeutic combination for targeting PPM1D-mutant DIPG tumors. IMPLICATIONS: The findings support the use of PARPi in combination with PPM1D inhibition against PPM1D-mutant DIPGs.


Asunto(s)
Aminopiridinas/farmacología , Neoplasias del Tronco Encefálico/genética , Glioma Pontino Intrínseco Difuso/genética , Dipéptidos/farmacología , Mutación , Ftalazinas/farmacología , Piperazinas/farmacología , Proteína Fosfatasa 2C/genética , Regulación Alostérica , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioma Pontino Intrínseco Difuso/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Fosfatasa 2C/antagonistas & inhibidores , Recombinasa Rad51/metabolismo
9.
Biomed Pharmacother ; 125: 109956, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32006900

RESUMEN

A greater understanding of factors causing cancer initiation, progression and evolution is of paramount importance. Among them, the serine/threonine phosphatase PPM1D, also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is emerging as an important oncoprotein due to its negative regulation on a number of crucial cancer suppressor pathways. Initially identified as a p53-regulated gene, PPM1D has been afterwards found amplified and more recently mutated in many human cancers such as breast cancer. The latest progress in this field further reveals that selective inhibition of PPM1D to delay tumor onset or reduce tumor burden represents a promising anti-cancer strategy. Here, we review the advances in the studies of the PPM1D activity and its relevance to various cancers, and recent progress in development of PPM1D inhibitors and discuss their potential application in cancer therapy. Consecutive research on PPM1D and its relationship with cancer is essential, as it ultimately contributes to the etiology and treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/metabolismo , Antineoplásicos/química , Estructura Molecular , Neoplasias/tratamiento farmacológico , Proteína Fosfatasa 2C/genética
10.
Sci Rep ; 9(1): 18795, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31827209

RESUMEN

ERBB2 amplification is a prognostic marker for aggressive tumors and a predictive marker for prolonged survival following treatment with HER2 inhibitors. We attempt to sub-group HER2+ tumors based on amplicon structures and co-amplified genes. We examined five HER2+ cell lines, three HER2+ xenographs and 57 HER2+ tumor tissues. ERBB2 amplification was analyzed using digital droplet PCR and low coverage whole genome sequencing. In some HER2+ tumors PPM1D, that encodes WIP1, is co-amplified. Cell lines were treated with HER2 and WIP1 inhibitors. We find that inverted duplication is the amplicon structure in the majority of HER2+ tumors. In patients suffering from an early stage disease the ERBB2 amplicon is composed of a single segment while in patients suffering from advanced cancer the amplicon is composed of several different segments. We find robust WIP1 inhibition in some HER2+ PPM1D amplified cell lines. Sub-grouping HER2+ tumors using low coverage whole genome sequencing identifies inverted duplications as the main amplicon structure and based on the number of segments, differentiates between local and advanced tumors. In addition, we found that we could determine if a tumor is a recurrent tumor or second primary tumor and identify co-amplified oncogenes that may serve as targets for therapy.


Asunto(s)
Amplificación de Genes , Neoplasias/clasificación , Receptor ErbB-2/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Línea Celular Tumoral , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Genes erbB-2 , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/genética , Reacción en Cadena de la Polimerasa , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/genética , Secuenciación Completa del Genoma , Adulto Joven
11.
Cells ; 8(10)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31619012

RESUMEN

Genotoxic stress triggers a combined action of DNA repair and cell cycle checkpoint pathways. Protein phosphatase 2C delta (referred to as WIP1) is involved in timely inactivation of DNA damage response by suppressing function of p53 and other targets at chromatin. Here we show that WIP1 promotes DNA repair through homologous recombination. Loss or inhibition of WIP1 delayed disappearance of the ionizing radiation-induced 53BP1 foci in S/G2 cells and promoted cell death. We identify breast cancer associated protein 1 (BRCA1) as interactor and substrate of WIP1 and demonstrate that WIP1 activity is needed for correct dynamics of BRCA1 recruitment to chromatin flanking the DNA lesion. In addition, WIP1 dephosphorylates 53BP1 at Threonine 543 that was previously implicated in mediating interaction with RIF1. Finally, we report that inhibition of WIP1 allowed accumulation of DNA damage in S/G2 cells and increased sensitivity of cancer cells to a poly-(ADP-ribose) polymerase inhibitor olaparib. We propose that inhibition of WIP1 may increase sensitivity of BRCA1-proficient cancer cells to olaparib.


Asunto(s)
Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatina/metabolismo , Daño del ADN/genética , Daño del ADN/fisiología , Reparación del ADN/genética , Reparación del ADN/fisiología , Resistencia a Antineoplásicos/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular , Células HEK293 , Recombinación Homóloga/genética , Humanos , Proteína Fosfatasa 2C/genética , Puntos de Control de la Fase S del Ciclo Celular , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
12.
J BUON ; 24(4): 1464-1469, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31646792

RESUMEN

PURPOSE: Accumulating reports have shown the oncogenic properties of PPMD1 (protein phosphatase, Mg2+/Mn2+ dependent 1D) in different cancer types. This study was undertaken to explore the role and therapeutic potential of PPM1D in colon cancer. METHODS: HT-29 colon cancer cell line was used in this study. Expression analysis of PPMD1 was performed by qRT-PCR. Cell viability was determined by CCK-8 assay. DAPI, acridin orange/ethidium bromide (AO/EB) and propidium iodide (PI)staining assays were used for apoptosis detection. Cell cycle analysis was performed by flow cytometry. Protein expression was determined by western blot analysis. RESULTS: The results showed that the expression of PPMD1 was significantly upregulated in colon cancer by 3.2 to 4.8 fold. Silencing of PPMD1 caused significant decline in the proliferation rate of the HT-29 colon cancer cells that was due to induction of apoptosis as evidenced by DAPI, AO/EB and PI staining. Annexin V/PI showed a significant increase in the percentage of apoptotic of HT-29 cells upon silencing of PPMD1. The induction of apoptosis was also accompanied by increase in Bax and decrease in Bcl-2 expression. PPMD1 silencing also resulted in arrest of the HT-29 cells in the G2/M phase of the cell cycle which was also associated with upsurge of p21 and p53 and depletion of cyclin B1 expression levels. PPMD1 silencing also caused decrease in the viability of the HT-29 cells which was concomitant with suppression of MMP-2 and MMP-9 expression. CONCLUSION: These findings suggest that PPMD1 has oncogenic properties in colon cancer and exhibit therapeutic implications in colon cancer treatment.


Asunto(s)
Biomarcadores de Tumor/genética , Proliferación Celular/genética , Neoplasias del Colon/genética , Proteína Fosfatasa 2C/genética , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Supervivencia Celular/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/genética , Células HT29 , Humanos , Proteína Fosfatasa 2C/antagonistas & inhibidores
13.
J Biol Chem ; 294(46): 17654-17668, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31481464

RESUMEN

WT P53-Induced Phosphatase 1 (WIP1) is a member of the magnesium-dependent serine/threonine protein phosphatase (PPM) family and is induced by P53 in response to DNA damage. In several human cancers, the WIP1 protein is overexpressed, which is generally associated with a worse prognosis. Although WIP1 is an attractive therapeutic target, no potent, selective, and bioactive small-molecule modulator with favorable pharmacokinetics has been reported. Phosphatase enzymes are among the most challenging targets for small molecules because of the difficulty of achieving both modulator selectivity and bioavailability. Another major obstacle has been the availability of robust and physiologically relevant phosphatase assays that are suitable for high-throughput screening. Here, we describe orthogonal biochemical WIP1 activity assays that utilize phosphopeptides from native WIP1 substrates. We optimized an MS assay to quantify the enzymatically dephosphorylated peptide reaction product in a 384-well format. Additionally, a red-shifted fluorescence assay was optimized in a 1,536-well format to enable real-time WIP1 activity measurements through the detection of the orthogonal reaction product, Pi We validated these two optimized assays by quantitative high-throughput screening against the National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection and used secondary assays to confirm and evaluate inhibitors identified in the primary screen. Five inhibitors were further tested with an orthogonal WIP1 activity assay and surface plasmon resonance binding studies. Our results validate the application of miniaturized physiologically relevant and orthogonal WIP1 activity assays to discover small-molecule modulators from high-throughput screens.


Asunto(s)
Activadores de Enzimas/química , Fosfopéptidos/química , Proteína Fosfatasa 2C/química , Bibliotecas de Moléculas Pequeñas/química , Activadores de Enzimas/aislamiento & purificación , Activadores de Enzimas/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteína Fosfatasa 2C/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/farmacología , Especificidad por Sustrato , Proteína p53 Supresora de Tumor/química
14.
J Hypertens ; 37(11): 2256-2268, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31136458

RESUMEN

OBJECTIVES: Vascular smooth muscle cell (VSMC) proliferation is a crucial cause of vascular neointima hyperplasia and restenosis, thus limiting the long-term efficacy of percutaneous vascular intervention. We explored the role of wild-type p53-induced phosphatase 1 (Wip1), a potent regulator of tumorigenesis and atherosclerosis, in VSMC proliferation and neointima hyperplasia. METHODS AND RESULTS: Animal model of vascular restenosis was established in wild type C57BL/6J and VSMC-specific Tuberous Sclerosis 1 (TSC1)-knockdown mice by wire injury. We observed increased protein levels of Wip1, phospho (p)-S6 Ribosomal Protein (S6), p-4EBP1 but decreased p-adenosine 5'-monophosphate-activated protein kinase (AMPK)α both in carotid artery at day 28 after injury and in VSMCs after 48 h of platelet derived growth factor-BB (PDGF-BB) treatment. By using hematoxylin-eosin staining, Ki-67 immunohistochemical staining, cell counting kit-8 assay and Ki-67 immunofluorescence staining, we found Wip1 antagonist GSK2830371 (GSK) or mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin both obviously reversed the neointima formation and VSMC proliferation induced by wire injury and PDGF-BB, respectively. GSK also reversed the increase in mRNA level of Collagen I after wire injury. However, GSK had no obvious effects on VSMC migration induced by PDGF-BB. Simultaneously, TSC1 knockdown as well as AMPK inhibition by Compound C abolished the vascular protective and anti-proliferative effects of Wip1 inhibition. Additionally, suppression of AMPK also reversed the declined mTORC1 activity by GSK. CONCLUSION: Wip1 promotes VSMC proliferation and neointima hyperplasia after wire injury via affecting AMPK/mTORC1 pathway.


Asunto(s)
Aminopiridinas/uso terapéutico , Dipéptidos/uso terapéutico , Miocitos del Músculo Liso/efectos de los fármacos , Neointima/prevención & control , Proteína Fosfatasa 2C/metabolismo , Lesiones del Sistema Vascular/enzimología , Proteínas Quinasas Activadas por AMP/metabolismo , Aminopiridinas/farmacología , Animales , Becaplermina , Arteria Carótida Común/patología , Proliferación Celular/efectos de los fármacos , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Hiperplasia , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular , Neointima/etiología , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Lesiones del Sistema Vascular/complicaciones
15.
Neuro Oncol ; 21(6): 786-799, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30852603

RESUMEN

BACKGROUND: Children with diffuse intrinsic pontine glioma (DIPG) succumb to disease within 2 years of diagnosis despite treatment with ionizing radiation (IR) and/or chemotherapy. Our aim was to determine the role of protein phosphatase, magnesium-dependent 1, delta (PPM1D) mutation, present in up to 25% of cases, in DIPG pathogenesis and treatment. METHODS: Using genetic and pharmacologic approaches, we assayed effects of PPM1D mutation on DIPG growth and murine survival. We assayed effects of targeting mutated PPM1D alone or with IR on signaling, cell cycle, proliferation, and apoptosis in patient-derived DIPG cells in vitro, in organotypic brain slices, and in vivo. RESULTS: PPM1D-mutated DIPG cell lines exhibited increased proliferation in vitro and in vivo, conferring reduced survival in orthotopically xenografted mice, through stabilization of truncated PPM1D protein and inactivation of DNA damage response (DDR) effectors p53 and H2A.X. PPM1D knockdown or treatment with PPM1D inhibitors suppressed growth of PPM1D-mutated DIPGs in vitro. Orthotopic xenografting of PPM1D short hairpin RNA-transduced or PPM1D inhibitor-treated, PPM1D-mutated DIPG cells into immunodeficient mice resulted in reduced tumor proliferation, increased apoptosis, and extended mouse survival. PPM1D inhibition had similar effects to IR alone on DIPG growth inhibition and augmented the anti-proliferative and pro-apoptotic effects of IR in PPM1D-mutated DIPG models. CONCLUSIONS: PPM1D mutations inactivate DDR and promote DIPG growth. Treatment with PPM1D inhibitors activated DDR pathways and enhanced the anti-proliferative and pro-apoptotic effects of IR in DIPG models. Our results support continued development of PPM1D inhibitors for phase I/II trials in children with DIPG.


Asunto(s)
Neoplasias del Tronco Encefálico/radioterapia , Daño del ADN/efectos de la radiación , Glioma Pontino Intrínseco Difuso/radioterapia , Proteínas Mutantes/antagonistas & inhibidores , Mutación , Proteína Fosfatasa 2C/antagonistas & inhibidores , Radiación Ionizante , Animales , Apoptosis , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/patología , Ciclo Celular , Movimiento Celular , Proliferación Celular , Glioma Pontino Intrínseco Difuso/genética , Glioma Pontino Intrínseco Difuso/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteína Fosfatasa 2C/genética , Proteína Fosfatasa 2C/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 14(2): e0212682, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30811466

RESUMEN

Obesity is a worldwide public health problem, which is associated with various severe diseases including diabetes, hypertension, atherosclerosis, and cancer. Recent studies have revealed that combination treatment of several different compounds using low doses is effective to reduce side effects. Thus, there is a need to develop an efficient inhibitor for reducing lipid droplets with a divergent target/pathway. Ser/Thr protein phosphatase PPM1D is involved in cellular metabolic processes and is a promising target for anti-obesity treatment. We have previously developed a potent and specific PPM1D inhibitor, SL-176. In this study, we demonstrated that significant reduction of lipid droplet formation in adipocytes by the PPM1D specific inhibitor, SL-176. Using Oil-red O staining and fluorescent imaging of lipid droplet, we found that treatment of SL-176 significantly suppressed lipid droplet formation of 3T3-L1 cells both in amount and in size. SL-176 also repressed mRNA and protein expression of PPARγ and C/EBPα, adipogenic markers, at nontoxic conditions. Thus, SL-176 is a unique and potent inhibitor of lipid droplet formation that acts via PPM1D, a novel target toward inhibiting adipocyte differentiation.


Asunto(s)
Adipocitos/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Gotas Lipídicas/efectos de los fármacos , Naftalenos/farmacología , Compuestos de Organosilicio/farmacología , Proteína Fosfatasa 2C/antagonistas & inhibidores , Células 3T3-L1 , Adipocitos/citología , Adipocitos/fisiología , Adipogénesis/efectos de los fármacos , Animales , Fármacos Antiobesidad/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ratones , Naftalenos/uso terapéutico , Obesidad/tratamiento farmacológico , Compuestos de Organosilicio/uso terapéutico , Proteína Fosfatasa 2C/metabolismo
17.
Antioxid Redox Signal ; 30(17): 1983-1998, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-29808718

RESUMEN

Aims: Epidemiologic evidence indicates that diabetes may increase risk of breast cancer (BC) and mortality in patients with cancer. The pathophysiological relationships between diabetes and cancer are not fully understood, and personalized treatments for diabetes-associated BC are urgently needed. Results: We observed that high glucose (HG), via activation of nuclear phosphatase PP2Cδ, suppresses p53 function, and consequently promotes BC cell proliferation, migration, and invasion. PP2Cδ expression is higher in tumor tissues from BC patients with hyperglycemia than those with normoglycemia. The mechanisms underlying HG stimulation of PP2Cδ involve classical/novel protein kinase-C (PKC) activation and GSK3ß phosphorylation. Reactive oxygen species (ROS)/NF-κB pathway also mediates HG induction of PP2Cδ. Furthermore, we identified a 1,5-diheteroarylpenta-1,4-dien-3-one (Compound 23, or C23) as a novel potent PP2Cδ inhibitor with a striking cytotoxicity on MCF-7 cells through cell-based screening assay for growth inhibition and activity of a group of curcumin mimics. Beside directly inhibiting PP2Cδ activity, C23 blocks HG induction of PP2Cδ expression via heat shock protein 27 (HSP27) induction and subsequent ablation of ROS/NF-κB activation. C23 can thus significantly block HG-triggered inhibition of p53 activity, leading to the inhibition of cancer cell proliferation, migration, and invasion. In addition, hyperglycemia promotes BC development in diabetic nude mice, and C23 inhibits the xenografted BC tumor growth. Conclusions and Innovation: Our findings elucidate mechanisms that may have contributed to diabetes-associated BC progression, and provide the first evidence to support the possible alternative therapeutic approach to BC patients with diabetes. Antioxid. Redox Signal. 30, 1983-1998.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Curcumina/farmacología , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Proteína Fosfatasa 2C/antagonistas & inhibidores , Acetilación , Animales , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Curcumina/análogos & derivados , Curcumina/química , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inhibidores Enzimáticos/química , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Hiperglucemia , Ratones , Modelos Moleculares , FN-kappa B/metabolismo , Fosforilación , Proteína Fosfatasa 2C/química , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Biochem ; 165(6): 471-477, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30576481

RESUMEN

The protein phosphatase PPM1D (Wip1) was originally identified as a p53 target product. Activation of PPM1D through various mechanism promotes the tumorigenic potential of various cancers by suppressing p53 and other DNA damage response proteins. New functions of PPM1D have recently been revealed in physiological processes such as cell differentiation. However, the regulatory mechanisms of signalling pathway to maintain stemness and induce cell differentiation are still unclear. Here we report that PPM1D modulates retinoic acid (RA) signalling. PPM1D knockdown resulted in decreased alkaline phosphatase activity of the human teratocarcinoma cell line NT2/D1. Inhibition of PPM1D-induced cell differentiation and decreased gene expression of the stem cell marker Oct-4 (POU5F1). RA-induced cell differentiation was promoted by reducing PPM1D activity. RA treatment elicited activation of the MEK-ERK pathway and induced rapid and transient activation of the extracellular signal-regulated kinase 1/2 (ERK-1/2). PPM1D dephosphorylated a phosphopeptide with the TEY motif in ERK-1/2 in vitro. Moreover, phosphorylation of ERK-1/2 was facilitated by PPM1D inhibition. Our study shows that PPM1D plays an important role in maintaining the undifferentiation state and a new function in RA-induced ERK regulation and cell differentiation.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Embrionario/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteína Fosfatasa 2C/antagonistas & inhibidores , Tretinoina/farmacología , Carcinoma Embrionario/metabolismo , Carcinoma Embrionario/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Fosfatasa 2C/metabolismo , Relación Estructura-Actividad
19.
Int J Cardiol ; 270: 228-236, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30220377

RESUMEN

BACKGROUND: Excessive cardiac fibrosis due to maladaptive remodeling leads to progression of cardiac dysfunction and is modulated by TGF-ß1-activated intracellular phospho-SMAD signaling effectors and transcription regulators. SMAD2/3 phosphorylation, regulated by protein-phosphatases, has been studied in different cell types, but its role in human ventricular fibroblasts (hVFs) is not defined as a target to reduce cytokine-mediated excessive fibrotic response and adverse cardiac remodeling. Statins are a class of drugs reported to reduce cardiac fibrosis, although underlying mechanisms are not completely understood. We aimed to assess whether simvastatin-mediated reduction in TGF-ß1-augmented profibrotic response involves reduction in phospho-SMAD2/3 owing to activation of protein-phosphatase in hVFs. METHODS AND RESULTS: Cultures of hVFs were used. Effect of simvastatin on TGF-ß1-treated hVF proliferation, cytotoxicity, myofibroblast differentiation/activation, profibrotic gene expression and protein-phosphatase activity was assessed. Simvastatin (1 µM) reduced effect of TGF-ß1 (5 ng/mL) on hVF proliferation, myofibroblast differentiation (reduced α-smooth muscle actin [α-SMA-expression]) and activation (decreased procollagen-peptide release). Simvastatin also reduced TGF-ß1-stimulated time-dependent increases in SMAD2/3 phosphorylation and nuclear translocation, mediated through catalytic activation of protein-phosphatases PPM1A and PP2A, which physically interact with SMAD2/3, thereby promoting their dephosphorylation. Effect of simvastatin on TGF-ß1-induced fibroblast activation was annulled by okadaic acid, an inhibitor of protein-phosphatase. CONCLUSIONS: This proof-of-concept study using an in vitro experimental cell culture model identifies the protective role of simvastatin against TGF-ß1-induced hVF transformation into activated myofibroblasts through activation of protein phosphatase, a novel target that can be therapeutically modulated to curb excessive cardiac fibrosis associated with maladaptive cardiac remodeling.


Asunto(s)
Fibroblastos/metabolismo , Proteína Fosfatasa 2C/metabolismo , Proteína Fosfatasa 2/metabolismo , Simvastatina/farmacología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/toxicidad , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Fibroblastos/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2C/antagonistas & inhibidores
20.
J Bacteriol ; 200(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30038048

RESUMEN

Chlamydiae are obligate intracellular Gram-negative bacterial pathogens that undergo an essential, but poorly understood, biphasic developmental cycle transitioning between the infectious elementary body and the replicative reticulate body. Ser/Thr/Tyr phosphorylation has been increasingly recognized for its role in regulating bacterial physiology. Chlamydia spp. encode two Hanks'-type kinases in addition to a type 2C protein phosphatase (PP2C; CppA) and appears capable of global protein phosphorylation. While these findings substantiate the importance of protein phosphorylation in Chlamydia, the physiological impact of protein phosphorylation remains enigmatic. In this study, we investigated the in vivo role of CppA by using recombinant protein point mutants and small-molecule inhibitors. Recombinant CppA (rCppA) amino acid point mutants based upon missense mutations identified in growth-deficient Chlamydia trachomatis strains exhibited reduced, but not a complete loss of, phosphatase activity toward p-nitrophenyl phosphate (pNPP) and phosphopeptides. To more directly explore the importance of CppA in chlamydial development, we implemented a chemical "knockout" approach using derivatives of 5,5'-methylenedisalicylic acid (MDSA). Several MDSA derivatives significantly reduced CppA activity in vitro and the growth of C. trachomatis L2, C. trachomatis D, and Chlamydia muridarum in a cell culture infection model. The inhibition of C. trachomatis L2 growth was more pronounced when treated at earlier infection time points, and the removal of the inhibitors after 12 h postinfection did not rescue progeny production. Our findings revealed that altered CppA activity reduces chlamydial growth and that CppA function is likely crucial for early differentiation events. Collectively, our findings further support the importance of the protein phosphorylation network in chlamydial development.IMPORTANCEChlamydia is a significant cause of disease in humans, including sexually transmitted infections, the ocular infection trachoma, and pneumonia. Despite the critical roles of protein phosphatases in bacterial physiology, their function in pathogenesis is less clear. Our findings demonstrate that CppA, a broad-specificity type 2C protein phosphatase (PP2C), is critical for chlamydial development and further substantiate reversible phosphorylation as a key regulatory mechanism in Chlamydia Additionally, our work highlights the potential of CppA to serve as a novel target for future therapeutic strategies and supports the feasibility of designing more potent PP2C phosphatase inhibitors for Chlamydia and other pathogenic bacteria.


Asunto(s)
Proteínas Bacterianas/genética , Chlamydia trachomatis/efectos de los fármacos , Chlamydia trachomatis/genética , Proteína Fosfatasa 2C/antagonistas & inhibidores , Proteína Fosfatasa 2C/genética , Salicilatos/farmacología , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Línea Celular , Regulación Bacteriana de la Expresión Génica , Células HeLa , Humanos , Ratones , Fosforilación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...