Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Plant Cell ; 34(2): 742-758, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-34865106

RESUMO

During moderate severity drought and low water potential (ψw) stress, poorly understood signaling mechanisms restrict both meristem cell division and subsequent cell expansion. We found that the Arabidopsis thaliana Clade E Growth-Regulating 2 (EGR2) protein phosphatase and Microtubule-Associated Stress Protein 1 (MASP1) differed in their stoichiometry of protein accumulation across the root meristem and had opposing effects on root meristem activity at low ψw. Ectopic MASP1 or EGR expression increased or decreased, respectively, root meristem size and root elongation during low ψw stress. This, along with the ability of phosphomimic MASP1 to overcome the EGR-mediated suppression of root meristem size and the observation that ectopic EGR expression had no effect on unstressed plants, indicated that during low ψw EGR activation and attenuation of MASP1 phosphorylation in their overlapping zone of expression determines root meristem size and activity. Ectopic EGR expression also decreased root cell size at low ψw. Conversely, both the egr1-1 egr2-1 and egr1-1 egr2-1 masp1-1 mutants had similarly increased root cell size but only egr1-1egr2-1 had increased cell division. These observations demonstrated that EGRs affect meristem activity via MASP1 but affect cell expansion via other mechanisms. Interestingly, EGR2 was highly expressed in the root cortex, a cell type important for growth regulation and environmental response.


Assuntos
Proteínas de Arabidopsis/fisiologia , Arabidopsis/fisiologia , Meristema/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Raízes de Plantas/fisiologia , Divisão Celular , Tamanho Celular , Desidratação , Secas , Regulação da Expressão Gênica de Plantas , Meristema/citologia , Células Vegetais , Plantas Geneticamente Modificadas , Proteína Fosfatase 2C/fisiologia
3.
Int J Mol Sci ; 22(1)2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33401385

RESUMO

Clade A Type 2C protein phosphatases (PP2CAs) negatively regulate abscisic acid (ABA) signaling and have diverse functions in plant development and in response to various stresses. In this study, we showed that overexpression of the rice ABA receptor OsPYL/RCAR3 reduces the growth retardation observed in plants exposed to osmotic stress. By contrast, overexpression of the OsPYL/RCAR3-interacting protein OsPP2C09 rendered plant growth more sensitive to osmotic stress. We tested whether OsPP2CAs activate an ABA-independent signaling cascade by transfecting rice protoplasts with luciferase reporters containing the drought-responsive element (DRE) or ABA-responsive element (ABRE). We observed that OsPP2CAs activated gene expression via the cis-acting drought-responsive element. In agreement with this observation, transcriptome analysis of plants overexpressing OsPP2C09 indicated that OsPP2C09 induces the expression of genes whose promoters contain DREs. Further analysis showed that OsPP2C09 interacts with DRE-binding (DREB) transcription factors and activates reporters containing DRE. We conclude that, through activating DRE-containing promoters, OsPP2C09 positively regulates the drought response regulon and activates an ABA-independent signaling pathway.


Assuntos
Oryza/enzimologia , Proteína Fosfatase 2C/metabolismo , Transdução de Sinais , Estresse Fisiológico , Ácido Abscísico/metabolismo , Secas , Regulação da Expressão Gênica de Plantas , Oryza/genética , Oryza/fisiologia , Pressão Osmótica , Proteínas de Plantas/metabolismo , Proteínas de Plantas/fisiologia , Proteína Fosfatase 2C/fisiologia
4.
Biochem Pharmacol ; 184: 114362, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33309518

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Terapia de Alvo Molecular/métodos , Proteína Fosfatase 2C/antagonistas & inibidores , Proteína Fosfatase 2C/fisiologia , Animais , Ciclo Celular , Dano ao DNA , Feminino , Fertilidade/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Masculino , Camundongos
5.
Reprod Fertil Dev ; 32(18): 1350-1356, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33287951

RESUMO

Wild-type p53-induced phosphatase 1 (WIP1) plays an oncogenic function by increasing cell proliferation in various cancer types. Deficiency in WIP1 expression leads to male infertility, possibly by impairing the blood-testis barrier and spermatogenesis. However, how WIP1 functions in the Sertoli cells to affect male reproduction remains unclear. Thus, in the present study we used a swine Sertoli cell line to investigate whether WIP1 regulated the proliferation of Sertoli cells to participate in male reproduction. The WIP1 inhibitor GSK2830371, WIP1-short interference (si) RNAs and an upstream microRNA (miR-16) were used to inhibit the expression of WIP1, after which the proliferation of swine Sertoli cells, P53 expression and the levels of P53 phosphorylation were determined. Inhibiting WIP1 expression suppressed swine Sertoli cell proliferation, increased P53 expression and increased levels of P53 phosphorylation. In addition, overexpression of miR-16 in swine Sertoli cells resulted in a decrease in WIP1 expression and increases in both P53 expression and P53 phosphorylation. Together, these findings suggest that WIP1 positively regulates the proliferation of swine Sertoli cells by inhibiting P53 phosphorylation, and the miR-16 is likely also involved by targeting WIP1.


Assuntos
Proliferação de Células/genética , Proteína Fosfatase 2C/fisiologia , Células de Sertoli/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Células Cultivadas , Regulação Enzimológica da Expressão Gênica , Masculino , MicroRNAs/fisiologia , Fosforilação , Proteína Fosfatase 2C/genética , Processamento de Proteína Pós-Traducional , Suínos , Proteína Supressora de Tumor p53/metabolismo
6.
Biochem Biophys Res Commun ; 533(4): 1309-1314, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33051059

RESUMO

Spatial learning and memory are typically assessed to evaluate hippocampus-dependent cognitive and memory functions in vivo. Protein phosphorylation and dephosphorylation by kinases and phosphatases play critical roles in spatial learning and memory. Here we report that the Wip1 phosphatase is essential for spatial learning, with knockout mice lacking Wip1 phosphatase exhibiting dysfunctional spatial cognition. Aberrant phosphorylation of the Wip1 substrates p38, ATM, and p53 were observed in the hippocampi of Wip1-/- mice, but only p38 inhibition reversed impairments in long-term potentiation in Wip1-knockout mice. p38 inhibition consistently ameliorated the spatial learning dysfunction caused by Wip1 deficiency. Our results demonstrate that deletion of Wip1 phosphatase impairs hippocampus-dependent spatial learning and memory, with aberrant downstream p38 phosphorylation involved in this process and providing a potential therapeutic target.


Assuntos
Memória , Proteína Fosfatase 2C/fisiologia , Aprendizagem Espacial , Animais , Hipocampo/enzimologia , Hipocampo/fisiologia , Potenciação de Longa Duração , Masculino , Camundongos Knockout , Teste do Labirinto Aquático de Morris , Fosforilação , Proteína Fosfatase 2C/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Cells ; 9(9)2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32927737

RESUMO

Genome integrity is protected by the cell-cycle checkpoints that prevent cell proliferation in the presence of DNA damage and allow time for DNA repair. The transient checkpoint arrest together with cellular senescence represent an intrinsic barrier to tumorigenesis. Tumor suppressor p53 is an integral part of the checkpoints and its inactivating mutations promote cancer growth. Protein phosphatase magnesium-dependent 1 (PPM1D) is a negative regulator of p53. Although its loss impairs recovery from the G2 checkpoint and promotes induction of senescence, amplification of the PPM1D locus or gain-of-function truncating mutations of PPM1D occur in various cancers. Here we used a transgenic mouse model carrying a truncating mutation in exon 6 of PPM1D (Ppm1dT). As with human cell lines, we found that the truncated PPM1D was present at high levels in the mouse thymus. Truncated PPM1D did not affect differentiation of T-cells in the thymus but it impaired their response to ionizing radiation (IR). Thymocytes in Ppm1dT/+ mice did not arrest in the checkpoint and continued to proliferate despite the presence of DNA damage. In addition, we observed a decreased level of apoptosis in the thymi of Ppm1dT/+ mice. Moreover, the frequency of the IR-induced T-cell lymphomas increased in Ppm1dT/+Trp53+/- mice resulting in decreased survival. We conclude that truncated PPM1D partially suppresses the p53 pathway in the mouse thymus and potentiates tumor formation under the condition of a partial loss of p53 function.


Assuntos
Apoptose , Linfoma/metabolismo , Proteína Fosfatase 2C/fisiologia , Timócitos/citologia , Timo , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular , Proliferação de Células , Dano ao DNA , Reparo do DNA , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Induzidas por Radiação/metabolismo , Radiação Ionizante , Timócitos/metabolismo , Timo/citologia , Timo/metabolismo
8.
Arthritis Rheumatol ; 72(5): 750-760, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31762216

RESUMO

OBJECTIVE: Increased protein phosphatase magnesium-dependent 1A (PPM1A) levels in patients with ankylosing spondylitis regulate osteoblast differentiation in bony ankylosis; however, the potential mechanisms that regulate osteoclast differentiation in relation to abnormal bone formation remain unclear. This study was undertaken to investigate the relationship of PPM1A to osteoclast differentiation by generating conditional gene-knockout (PPM1Afl/fl ;LysM-Cre) mice and evaluating their bone phenotype. METHODS: The bone phenotypes of LysM-Cre mice (n = 6) and PPM1Afl/fl ;LysM-Cre mice (n = 6) were assessed by micro-computed tomography. Osteoclast differentiation was induced by culturing bone marrow-derived macrophages in the presence of RANKL and macrophage colony-stimulating factor (M-CSF), and was evaluated by counting tartrate-resistant acid phosphatase-positive multinucleated cells. Levels of messenger RNA for PPM1A, RANK, and osteoclast-specific genes were examined by real-time quantitative polymerase chain reaction, and protein levels were determined by Western blotting. Surface RANK expression was analyzed by fluorescence flow cytometry. RESULTS: The PPM1Afl/fl ;LysM-Cre mice displayed reduced bone mass (P < 0.001) and increased osteoclast differentiation (P < 0.001) and osteoclast-specific gene expression (P < 0.05) compared with their LysM-Cre littermates. Mechanistically, reduced PPM1A function in osteoclast precursors in PPM1Afl/fl ;LysM-Cre mice induced osteoclast lineage commitment by up-regulating RANK expression (P < 0.01) via p38 MAPK activation in response to M-CSF. PPM1A expression in macrophages was decreased by Toll-like receptor 4 activation (P < 0.05). The Ankylosing Spondylitis Disease Activity Score was negatively correlated with the expression of PPM1A in peripheral blood mononuclear cells from patients with axial spondyloarthritis (SpA) (γ = -0.7072, P < 0.0001). CONCLUSION: The loss of PPM1A function in osteoclast precursors driven by inflammatory signals contributes to osteoclast lineage commitment and differentiation by elevating RANK expression, reflecting a potential role of PPM1A in dynamic bone metabolism in axial SpA.


Assuntos
Diferenciação Celular , Osteoclastos/fisiologia , Proteína Fosfatase 2C/fisiologia , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Ligante RANK/fisiologia
9.
Brain Dev ; 41(6): 538-541, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30795918

RESUMO

PPM1D truncating mutations in the last and penultimate exons of the gene have been associated with intellectual disability (ID) syndrome. Only 15 affected patients to-date have been reported with mild-to-severe ID, autistic behavior, anxiety and dysmorphic features. Here, we describe the clinical characteristics and underlying genetics of two unrelated girls with moderate developmental delay and dysmorphic features associated with novel mutations in PPM1D exon 5. The dysmorphic features demonstrated by these two patients are consistent with previously reported patients, including broad forehead, thin upper lip, brachydactyly, and hypoplastic nails. We identified a de novo PPM1D mutation in exon 5 of each patient (c.1250_1251insACCA p.V419Tfs*16 and c.1256_1257insCAAG p.S421Qfs*14) by panel sequencing for 4,813 disease-related genes. Both patients also had frameshift mutations (at different positions) that resulted in the same estimated termination codon at 434. These additional reports add to the growing literature on PPM1D-associated ID syndrome and help delineate the clinical phenotype and genetic basis.


Assuntos
Deficiência Intelectual/genética , Proteína Fosfatase 2C/genética , Proteína Fosfatase 2C/fisiologia , Criança , Pré-Escolar , Deficiências do Desenvolvimento/genética , Éxons/genética , Feminino , Mutação da Fase de Leitura/genética , Genótipo , Humanos , Deficiência Intelectual/metabolismo , Mutação/genética , Fenótipo , Sequenciamento do Exoma/métodos
10.
Inflammation ; 42(3): 1004-1014, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30684253

RESUMO

Protein phosphatase 2A (PP2A) is one main serine/threonine phosphatase in eukaryotes, and its activation changes have been linked to modulation of numerous pathological processes, such as cancer, inflammation, fibrosis, and neurodegenerative diseases. Acute respiratory distress syndrome (ARDS), the major cause of respiratory failure, remains with limited therapies available up to now. Alveolar macrophages (AMs) are essential to innate immunity and host defense, participating in the pathogenesis of ARDS. As a result, AMs are considered as a potential therapeutic target for ARDS. In our study, we firstly found that PP2A activity was significantly decreased in the lipopolysaccharide (LPS)-stimulated AMs. Furthermore, adoptive transfer of AMs with enhanced PP2A enzyme activity that was improved by C2-ceramide prior to LPS exposure alleviated acute lung inflammation. Conversely, AM-specific ablation of PP2ACα exacerbated inflammatory responses to LPS. Mechanistically, PP2ACα negatively regulates LPS-induced cytokine secretion of AMs by NF-κB and MAPK pathways. Together, these findings provide the evidence to guide the development of novel therapeutic options targeting PP2ACα for ARDS/acute lung injury.


Assuntos
Macrófagos Alveolares/química , Proteína Fosfatase 2C/fisiologia , Síndrome do Desconforto Respiratório/prevenção & controle , Animais , Citocinas/metabolismo , Humanos , Lipopolissacarídeos , Sistema de Sinalização das MAP Quinases , Macrófagos Alveolares/imunologia , NF-kappa B/metabolismo , Fatores de Proteção , Proteína Fosfatase 2C/análise , Proteína Fosfatase 2C/farmacologia , Síndrome do Desconforto Respiratório/induzido quimicamente
11.
EMBO J ; 38(1)2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30429206

RESUMO

OST1 (open stomata 1) protein kinase plays a central role in regulating freezing tolerance in Arabidopsis; however, the mechanism underlying cold activation of OST1 remains unknown. Here, we report that a plasma membrane-localized clade-E growth-regulating 2 (EGR2) phosphatase interacts with OST1 and inhibits OST1 activity under normal conditions. EGR2 is N-myristoylated by N-myristoyltransferase NMT1 at 22°C, which is important for its interaction with OST1. Moreover, myristoylation of EGR2 is required for its function in plant freezing tolerance. Under cold stress, the interaction of EGR2 and NMT1 is attenuated, leading to the suppression of EGR2 myristoylation in plants. Plant newly synthesized unmyristoylated EGR2 has decreased binding ability to OST1 and also interferes with the EGR2-OST1 interaction under cold stress. Consequently, the EGR2-mediated inhibition of OST1 activity is released. Consistently, mutations of EGRs cause plant tolerance to freezing, whereas overexpression of EGR2 exhibits decreased freezing tolerance. This study thus unravels a molecular mechanism underlying cold activation of OST1 by membrane-localized EGR2 and suggests that a myristoyl switch on EGR2 helps plants to adapt to cold stress.


Assuntos
Aclimatação , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/fisiologia , Arabidopsis , Temperatura Baixa/efeitos adversos , Proteínas Quinases/metabolismo , Proteína Fosfatase 2C/fisiologia , Aclimatação/genética , Arabidopsis/enzimologia , Arabidopsis/genética , Arabidopsis/crescimento & desenvolvimento , Arabidopsis/metabolismo , Membrana Celular/metabolismo , Resposta ao Choque Frio/genética , Resposta ao Choque Frio/fisiologia , Ativação Enzimática/genética , Ácidos Graxos Monoinsaturados/metabolismo , Congelamento , Regulação da Expressão Gênica de Plantas , Fosforilação , Plantas Geneticamente Modificadas , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais
12.
IET Syst Biol ; 12(1): 26-38, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29337287

RESUMO

This study proposes a two-dimensional (2D) oscillator model of p53 network, which is derived via reducing the multidimensional two-phase dynamics model into a model of ataxia telangiectasia mutated (ATM) and Wip1 variables, and studies the impact of p53-regulators on cell fate decision. First, the authors identify a 6D core oscillator module, then reduce this module into a 2D oscillator model while preserving the qualitative behaviours. The introduced 2D model is shown to be an excitable relaxation oscillator. This oscillator provides a mechanism that leads diverse modes underpinning cell fate, each corresponding to a cell state. To investigate the effects of p53 inhibitors and the intrinsic time delay of Wip1 on the characteristics of oscillations, they introduce also a delay differential equation version of the 2D oscillator. They observe that the suppression of p53 inhibitors decreases the amplitudes of p53 oscillation, though the suppression increases the sustained level of p53. They identify Wip1 and P53DINP1 as possible targets for cancer therapies considering their impact on the oscillator, supported by biological findings. They model some mutations as critical changes of the phase space characteristics. Possible cancer therapeutic strategies are then proposed for preventing these mutations' effects using the phase space approach.


Assuntos
Raios gama , Modelos Teóricos , Proteína Supressora de Tumor p53 , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/fisiologia , Proteínas de Choque Térmico/fisiologia , Neoplasias/metabolismo , Neoplasias/terapia , Proteína Fosfatase 2C/fisiologia , Proteína Supressora de Tumor p53/fisiologia
13.
Mol Immunol ; 93: 31-37, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29128669

RESUMO

The blood brain barrier (BBB) is a diffusion barrier that maintains the brain environment. Wip1 is a nuclear phosphatase induced by many factors and involved in various stresses, tumorigenesis, organismal aging, and neurogenesis. Wip1's role in BBB integrity has not been thoroughly investigated. The purpose of the present study was to investigate the effect and mechanism of Wip1 on lipopolysaccharide (LPS)-induced BBB dysfunction and inflammation in an in vitro BBB model. The in vitro BBB model was established by co-culturing human brain-microvascular endothelial cells and human astrocytes and then exposing them to 1µg/ml LPS for 6, 12, 18, 24, and 48h. Wip1 expression was significantly elevated by LPS treatment. Knockdown of Wip1 aggravated the increased permeability and decreased transepithelial electrical resistance, protein expression of ZO-1, and occludin induced by LPS. Wip1 silencing augmented the elevated inflammatory cytokines TNF-α, IL-1ß, IL-12, and IL-6 of the BBB induced by LPS, whereas overexpression of Wip1 showed a contrary effect. Sonic hedgehog signaling (SHH) was activated by Wip1 overexpression and inhibited by Wip1 silencing. Additionally, activating or inhibiting the SHH pathway by purmorphamine or cyclopamine, respectively, abolished the Wip1-induced changes in transepithelial electrical resistance and permeability and inflammatory responses in the LPS-injured BBB model. Our results demonstrate that Wip1 may protect the BBB against LPS-induced integrity disruption and inflammatory response through the SHH signaling pathway.


Assuntos
Barreira Hematoencefálica/fisiologia , Encefalite/fisiopatologia , Células Endoteliais/efeitos dos fármacos , Proteínas Hedgehog/fisiologia , Lipopolissacarídeos/toxicidade , Proteína Fosfatase 2C/fisiologia , Transdução de Sinais/efeitos dos fármacos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Encefalite/induzido quimicamente , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Técnicas de Silenciamento de Genes , Humanos , Técnicas In Vitro , Morfolinas/farmacologia , Permeabilidade/efeitos dos fármacos , Proteína Fosfatase 2C/antagonistas & inibidores , Purinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/metabolismo , Alcaloides de Veratrum/farmacologia
14.
Neurosci Bull ; 33(3): 292-298, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28097612

RESUMO

The hypobaric hypoxic environment in high-altitude areas often aggravates the severity of inflammation and induces brain injury as a consequence. However, the critical genes regulating this process remain largely unknown. The phosphatase wild-type p53-induced phosphatase 1 (WIP1) plays important roles in various physiological and pathological processes, including the regulation of inflammation in normoxia, but its functions in hypoxic inflammation-induced brain injury remain unclear. Here, we established a mouse model of this type of injury and found that WIP1 deficiency augmented the release of inflammatory cytokines in the peripheral circulation and brain tissue, increased the numbers of activated microglia/macrophages in the brain, aggravated cerebral histological lesions, and exacerbated the impairment of motor and cognitive abilities. Collectively, these results provide the first in vivo evidence that WIP1 is a critical neuroprotector against hypoxic inflammation-induced brain injury.


Assuntos
Doença da Altitude , Lesões Encefálicas , Hipóxia , Inflamação , Neuroproteção/fisiologia , Proteína Fosfatase 2C/fisiologia , Doença da Altitude/complicações , Doença da Altitude/imunologia , Doença da Altitude/metabolismo , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças , Hipóxia/complicações , Hipóxia/imunologia , Hipóxia/metabolismo , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Fosfatase 2C/deficiência
15.
Oncotarget ; 7(43): 69625-69637, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27626308

RESUMO

PPM1D is a serine/threonine phosphatase that negatively regulates key DNA damage response proteins, such as p53, p38 MAPK, histone H2A.X, and ATM. We investigated the pathophysiological significance of PPM1D and its therapeutic targeting by the novel PPM1D inhibitor GSK2830371 in mantle cell lymphoma (MCL). Oncomine-based analyses indicated increased PPM1D mRNA levels in MCL cells compared with their normal counterpart cells. Higher PPM1D expression was associated with higher expression of the proliferation gene signature and poorer prognosis in patients. Eight MCL (three p53 wild-type and five mutant) cell lines were exposed to GSK2830371. GSK2830371 inhibited the cell growth, being prominent in p53 wild-type cells. GSK2830371 induced apoptosis in sensitive cells, as evidenced by induction of phosphatidylserine externalization and loss of mitochondrial membrane potential. p53 knockdown de-sensitized cell sensitivity. GSK2830371 increased the levels of total and Ser15-phosphorylated p53, and p53 targets p21 and PUMA. GSK2830371 and the MDM2 inhibitor Nutlin-3a acted synergistically in p53 wild-type cells. Interestingly, GSK2830371 sensitized MCL cells to bortezomib and doxorubicin in p53 wild-type and mutant cells; p38 signaling appeared to be involved in the GSK2830371/bortezomib lethality. PPM1D inhibition may represent a novel therapeutic strategy for MCL, which can be exploited in combination therapeutic strategies for MCL.


Assuntos
Aminopiridinas/farmacologia , Dipeptídeos/farmacologia , Linfoma de Célula do Manto/tratamento farmacológico , Proteína Fosfatase 2C/fisiologia , Transdução de Sinais/fisiologia , Aminopiridinas/uso terapêutico , Apoptose/efeitos dos fármacos , Bortezomib/farmacologia , Linhagem Celular Tumoral , Dipeptídeos/uso terapêutico , Doxorrubicina/farmacologia , Genes p53/fisiologia , Humanos , Imidazóis/farmacologia , Linfoma de Célula do Manto/mortalidade , Linfoma de Célula do Manto/patologia , Fosforilação , Piperazinas/farmacologia , Prognóstico , Proteína Fosfatase 2C/antagonistas & inibidores
16.
Brain Tumor Pathol ; 33(3): 191-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26942600

RESUMO

The aim of our study was to clarify the expression and gene copy number levels of protein phosphatase 1D magnesium-dependent, delta isoform (PPM1D), which is thought to be a regulator of the p53 protein in meningiomas of all three different WHO grades. Genomic DNA and mRNA were extracted from frozen tissues of meningiomas (WHO grade I, 20 cases; grade II, 17 cases; grade III, 20 cases). For analysis of the mRNA expression and gene dosage level of PPM1D, semiquantitative duplex RT-PCR, real-time RT-PCR, and semiquantitative duplex PCR were performed. We also analyzed several genes which locate near PPM1D in the genomic locus 17q22-24 using semiquantitative duplex RT-PCR. We found that the mean mRNA expression of PPM1D is higher in WHO grade II and III meningiomas than in grade I tumors. This finding is accompanied by moderate gene dosage increases for PPM1D in meningiomas of higher grades. Other genes located in the vicinity of PPM1D also showed mRNA overexpression in single meningioma cases. For these genes, however, no significant expression differences between meningioma grades could be observed. Thus, PPM1D in the chromosomal location 17q22-24 might be the most relevant candidate gene with respect to a potential functional implication in meningioma progression.


Assuntos
Dosagem de Genes/genética , Regulação Neoplásica da Expressão Gênica/genética , Expressão Gênica , Neoplasias Meníngeas/genética , Meningioma/genética , Proteína Fosfatase 2C/genética , Proteína Supressora de Tumor p53/genética , Cromossomos Humanos Par 17/genética , DNA de Neoplasias/genética , Progressão da Doença , Feminino , Humanos , Masculino , Neoplasias Meníngeas/patologia , Meningioma/patologia , Estadiamento de Neoplasias , Proteína Fosfatase 2C/fisiologia , RNA Neoplásico/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...