Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Clin Invest ; 131(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34596056

RESUMO

Enhanced signaling via RTKs in pulmonary hypertension (PH) impedes current treatment options because it perpetuates proliferation and apoptosis resistance of pulmonary arterial smooth muscle cells (PASMCs). Here, we demonstrated hyperphosphorylation of multiple RTKs in diseased human vessels and increased activation of their common downstream effector phosphatidylinositol 3'-kinase (PI3K), which thus emerged as an attractive therapeutic target. Systematic characterization of class IA catalytic PI3K isoforms identified p110α as the key regulator of pathogenic signaling pathways and PASMC responses (proliferation, migration, survival) downstream of multiple RTKs. Smooth muscle cell-specific genetic ablation or pharmacological inhibition of p110α prevented onset and progression of pulmonary hypertension (PH) as well as right heart hypertrophy in vivo and even reversed established vascular remodeling and PH in various animal models. These effects were attributable to both inhibition of vascular proliferation and induction of apoptosis. Since this pathway is abundantly activated in human disease, p110α represents a central target in PH.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Hipertensão Pulmonar/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Adulto , Animais , Células Cultivadas , Humanos , Hipertensão Pulmonar/etiologia , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
2.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34507989

RESUMO

The phosphoinositide 3-kinase regulatory subunit p85α is a key regulator of kinase signaling and is frequently mutated in cancers. In the present study, we showed that in addition to weakening the inhibitory interaction between p85α and p110α, a group of driver mutations in the p85α N-terminal SH2 domain activated EGFR, HER2, HER3, c-Met, and IGF-1R in a p110α-independent manner. Cancer cells expressing these mutations exhibited the activation of p110α and the AKT pathway. Interestingly, the activation of EGFR, HER2, and c-Met was attributed to the ability of driver mutations to inhibit HER3 ubiquitination and degradation. The resulting increase in HER3 protein levels promoted its heterodimerization with EGFR, HER2, and c-Met, as well as the allosteric activation of these dimerized partners; however, HER3 silencing abolished this transactivation. Accordingly, inhibitors of either AKT or the HER family reduced the oncogenicity of driver mutations. The combination of these inhibitors resulted in marked synergy. Taken together, our findings provide mechanistic insights and suggest therapeutic strategies targeting a class of recurrent p85α mutations.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Domínio Catalítico/genética , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Células HCT116 , Humanos , Mutação , Neoplasias/genética , Fosfatidilinositol 3-Quinases/metabolismo , Domínios Proteicos/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais , Domínios de Homologia de src
3.
Pharmacol Res ; 165: 105415, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33412279

RESUMO

Genetically modified animal studies have revealed specific expression patterns and unequivocal roles of class I PI3K isoenzymes. PI3K(p110α), a catalytic subunit of class I PI3Ks is ubiquitously expressed and is well characterised in the cardiovascular system. Given that genetic inhibition of PI3K(p110α) causes lethal phenotype embryonically, the catalytic subunit is critically important in housekeeping and biological processes. A growing number of studies underpin crucial roles of PI3K(p110α) in cell survival, proliferation, hypertrophy and arrhythmogenesis. While the studies provide great insights, the precise mechanisms involved in PI3K(p110α) hypofunction and atrial fibrillation (AF) are not fully known. AF is a well recognised clinical problem with significant management limitations. In this translational review, we attempted a narration of PI3K(p110α) hypofunction in the molecular basis of AF pathophysiology. We sought to cautiously highlight the relevance of this molecule in the therapeutic approaches for AF management per se (i.e without conditions associate with cell proliferation, like cancer), and in mitigating effects of clinical risk factors in atrial substrate formation leading to AF progression. We also considered PI3K(p110α) in AF gene association, with the aim of identifying mechanistic links between the ever increasingly well-defined genetic loci (regions and genes) and AF. Such mechanisms will aid in identifying new drug targets for arrhythmogenic substrate and AF.


Assuntos
Antiarrítmicos/uso terapêutico , Fibrilação Atrial/fisiopatologia , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Animais , Antiarrítmicos/farmacologia , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/etiologia , Classe Ia de Fosfatidilinositol 3-Quinase/efeitos dos fármacos , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Humanos
4.
FASEB J ; 34(9): 12521-12532, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32744782

RESUMO

Class Ia phosphoinositide 3-kinases (PI3K) are critical mediators of insulin and growth factor action. We have demonstrated that the p85α regulatory subunit of PI3K modulates the unfolded protein response (UPR) by interacting with and regulating the nuclear translocation of XBP-1s, a transcription factor essential for the UPR. We now show that PI3K activity is required for full activation of the UPR. Pharmacological inhibition of PI3K in cells blunts the ER stress-dependent phosphorylation of IRE1α and PERK, decreases induction of ATF4, CHOP, and XBP-1 and upregulates UPR target genes. Cells expressing a human p85α mutant (R649W) previously shown to inhibit PI3K, exhibit decreased activation of IRE1α and PERK and reduced induction of CHOP and ATF4. Pharmacological inhibition of PI3K, overexpression of a mutant of p85α that lacks the ability to interact with the p110α catalytic subunit (∆p85α) or expression of mutant p85α (R649W) in vivo, decreased UPR-dependent induction of ER stress response genes. Acute tunicamycin treatment of R649W+/- mice revealed reduced induction of UPR target genes in adipose tissue, whereas chronic tunicamycin exposure caused sustained increases in UPR target genes in adipose tissue. Finally, R649W+/- cells exhibited a dramatic resistance to ER stress-dependent apoptosis. These data suggest that PI3K pathway dysfunction causes ER stress that may drive the pathogenesis of several diseases including Type 2 diabetes and various cancers.


Assuntos
Tecido Adiposo/metabolismo , Apoptose , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Estresse do Retículo Endoplasmático , Resposta a Proteínas não Dobradas , Tecido Adiposo/citologia , Animais , Linhagem Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 1 de Ligação a X-Box/metabolismo
5.
Oncotarget ; 7(17): 23096-105, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27049833

RESUMO

Drug resistance is a growing problem that necessitates new strategies to combat pathogens. Neutrophil phagocytosis and production of intracellular ROS, in particular, has been shown to cooperate with antibiotics in the killing of microbes. This study tested the hypothesis that p85α, the regulatory subunit of PI3K, regulates production of intracellular ROS. Genetic knockout of p85α in mice caused decreased expression of catalytic subunits p110α, p110ß, and p110δ, but did not change expression levels of the NADPH oxidase complex subunits p67phox, p47phox, and p40phox. When p85α, p55α, and p50α (all encoded by Pik3r1) were deleted, there was an increase in intracellular ROS with no change in phagocytosis in response to both Fcγ receptor and complement receptor stimulation. Furthermore, the increased intracellular ROS correlated with significantly improved neutrophil killing of both methicillin-susceptible and methicillin-resistant S. aureus. Our findings suggest inhibition of p85α as novel approach to improving the clearance of resistant pathogens.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Camundongos , Camundongos Knockout , Transdução de Sinais
6.
J Neurosci ; 36(15): 4259-75, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-27076424

RESUMO

Growth of intact axons of noninjured neurons, often termed collateral sprouting, contributes to both adaptive and pathological plasticity in the adult nervous system, but the intracellular factors controlling this growth are largely unknown. An automated functional assay of genes regulated in sensory neurons from the rat in vivo spared dermatome model of collateral sprouting identified the adaptor protein CD2-associated protein (CD2AP; human CMS) as a positive regulator of axon growth. In non-neuronal cells, CD2AP, like other adaptor proteins, functions to selectively control the spatial/temporal assembly of multiprotein complexes that transmit intracellular signals. Although CD2AP polymorphisms are associated with increased risk of late-onset Alzheimer's disease, its role in axon growth is unknown. Assessments of neurite arbor structure in vitro revealed CD2AP overexpression, and siRNA-mediated knockdown, modulated (1) neurite length, (2) neurite complexity, and (3) growth cone filopodia number, in accordance with CD2AP expression levels. We show, for the first time, that CD2AP forms a novel multiprotein complex with the NGF receptor TrkA and the PI3K regulatory subunit p85, with the degree of TrkA:p85 association positively regulated by CD2AP levels. CD2AP also regulates NGF signaling through AKT, but not ERK, and regulates long-range signaling though TrkA(+)/RAB5(+) signaling endosomes. CD2AP mRNA and protein levels were increased in neurons during collateral sprouting but decreased following injury, suggesting that, although typically considered together, these two adult axonal growth processes are fundamentally different. These data position CD2AP as a major intracellular signaling molecule coordinating NGF signaling to regulate collateral sprouting and structural plasticity of intact adult axons. SIGNIFICANCE STATEMENT: Growth of noninjured axons in the adult nervous system contributes to adaptive and maladaptive plasticity, and dysfunction of this process may contribute to neurologic pathologies. Functional screening of genes regulated during growth of noninjured axons revealed CD2AP as a positive regulator of axon outgrowth. A novel association of CD2AP with TrkA and p85 suggests a distinct intracellular signaling pathway regulating growth of noninjured axons. This may also represent a novel mechanism of generating specificity in multifunctional NGF signaling. Divergent regulation of CD2AP in different axon growth conditions suggests that separate mechanisms exist for different modes of axon growth. CD2AP is the first signaling molecule associated with adult sensory axonal collateral sprouting, and this association may offer new insights for NGF/TrkA-related Alzheimer's disease mechanisms.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Axônios/fisiologia , Proteínas do Citoesqueleto/fisiologia , Fatores de Crescimento Neural/fisiologia , Plasticidade Neuronal/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Diferenciação Celular/genética , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Proteínas do Citoesqueleto/genética , Endossomos/metabolismo , Feminino , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/genética , Pseudópodes/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptor trkA/fisiologia , Transdução de Sinais/genética
7.
Eur J Gynaecol Oncol ; 36(4): 389-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26390688

RESUMO

OBJECTIVE: To investigate the expression of PI3K-p110α, pAkt, PTEN, the signaling molecules from PI3K/Akt signaling pathway, DJ-1, an oncoprotein and HSP90a, a molecular chaperone, and their correlation in uterine cervical neoplasia, in order to elucidate their role in cervical carcinogenesis. MATERIALS AND METHODS: Using immunohistochemistry, the authors analyzed the expression of PI3K-p110α, pAkt, PTEN, DJ-1 and HSP90α, and their correlation in ten normal tissues, cervical intraepithelial neoplasia (CIN) including 30 CIN1 and 31 CIN3, and 33 cases of invasive squamous cell carcinoma (SCC). RESULTS: The expression of all proteins significantly increased in CIN3 compared to CIN1, and only the expression of PI3K-p110α significantly increased in invasive SCC compared to CIN3. There was a significant positive correlation between the expression of PI3K-p110α and DJ-1, as well as PI3K-p110α and pAkt in CIN3 and invasive SCC. CONCLUSION: Overexpression of PI3K-p110α is associated with progression of uterine cervical neoplasia, and the expression of pAkt and DJ-1 is positively correlated with PI3K-p110α expression in this process.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Neoplasias do Colo do Útero/patologia , Classe Ia de Fosfatidilinositol 3-Quinase/análise , Progressão da Doença , Feminino , Proteínas de Choque Térmico HSP90/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/análise , Proteínas Oncogênicas/análise , PTEN Fosfo-Hidrolase/análise , Proteína Desglicase DJ-1 , Proteínas Proto-Oncogênicas c-akt/análise
8.
FEBS J ; 281(20): 4557-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25205393

RESUMO

Mammary gland involution involves a process that includes one of the most dramatic examples of cell death in an adult mammalian organism. We have previously shown that signal transducer and activator of transcription 3 (Stat3) regulates a lysosomal pathway of cell death in the first 48 h of involution and induces lysosome leakiness in mammary epithelial cells. Interestingly, Stat3 is associated also with the striking induction of autophagy that occurs concomitantly with cell death, presumably as a transient survival mechanism. The phosphatidylinositol 3-kinase regulatory subunits p55α and p50α are dramatically and specifically upregulated at the transcriptional level by Stat3 at the onset of involution. We show here that ablation of either Stat3 or p55α/p50α in vivo affects autophagy during involution. We used two different cell culture models (normal mammary epithelial cells and mouse embryonic fibroblasts) to further investigate the role of p55α/p50α in autophagy regulation. Our results demonstrate a direct role for p55α/p50α as inhibitors of autophagy mediated by p85α. Thus, Stat3 and its downstream targets p55α/p50α are key regulators of the balance between autophagy and cell death in vivo.


Assuntos
Autofagia , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Embrião de Mamíferos/patologia , Glândulas Mamárias Animais/patologia , Fator de Transcrição STAT3/fisiologia , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Técnicas Imunoenzimáticas , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Hepatology ; 59(5): 1779-90, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24806753

RESUMO

UNLABELLED: FAM3A belongs to a novel cytokine-like gene family, and its physiological role remains largely unknown. In our study, we found a marked reduction of FAM3A expression in the livers of db/db and high-fat diet (HFD)-induced diabetic mice. Hepatic overexpression of FAM3A markedly attenuated hyperglycemia, insulin resistance, and fatty liver with increased Akt (pAkt) signaling and repressed gluconeogenesis and lipogenesis in the livers of those mice. In contrast, small interfering RNA (siRNA)-mediated knockdown of hepatic FAM3A resulted in hyperglycemia with reduced pAkt levels and increased gluconeogenesis and lipogenesis in the livers of C57BL/6 mice. In vitro study revealed that FAM3A was mainly localized in the mitochondria, where it increases adenosine triphosphate (ATP) production and secretion in cultured hepatocytes. FAM3A activated Akt through the p110α catalytic subunit of PI3K in an insulin-independent manner. Blockade of P2 ATP receptors or downstream phospholipase C (PLC) and IP3R and removal of medium calcium all significantly reduced FAM3A-induced increase in cytosolic free Ca(2+) levels and attenuated FAM3A-mediated PI3K/Akt activation. Moreover, FAM3A-induced Akt activation was completely abolished by the inhibition of calmodulin (CaM). CONCLUSION: FAM3A plays crucial roles in the regulation of glucose and lipid metabolism in the liver, where it activates the PI3K-Akt signaling pathway by way of a Ca(2+) /CaM-dependent mechanism. Up-regulating hepatic FAM3A expression may represent an attractive means for the treatment of insulin resistance, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD).


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Citocinas/fisiologia , Gluconeogênese , Lipogênese , Fígado/metabolismo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Diabetes Mellitus Tipo 2/complicações , Dieta Hiperlipídica , Fígado Gorduroso/etiologia , Células Hep G2 , Humanos , Hiperglicemia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2/fisiologia
10.
Tumour Biol ; 35(8): 7853-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24819169

RESUMO

A post-transcriptional pathway by which TGF-ß modulates expression of specific proteins, Disabled-2 (Dab2) and Interleukin-like EMT Inducer (ILEI), inherent to epithelial to mesenchymal transition (EMT) in murine epithelial cells through Akt2-mediated phosphorylation of poly r(C) binding protein (PCBP1), has been previously elucidated. The aims of the current study were to determine if the same mechanism is operative in the non-small cell lung cancer (NSCLC) cell line, A549, and to delineate the underlying mechanism. Steady-state transcript and protein expression levels of Dab2 and ILEI were examined in A549 cells treated with TGF-ß for up to 48 h. Induction of translational de-repression in this model was quantified by polysomal fractionation followed by qRT-PCR. The underlying mechanism of isoform-specific activation of Akt2 was elucidated through a combination of co-immunoprecipitation studies. TGF-ß induced EMT in A549 cells concomitant with translational upregulation of Dab2 and ILEI proteins through isoform-specific activation of Akt2 followed by phosphorylation of PCBP1 at serine-43. Our experiments further elucidated that the adaptor protein SchA is phosphorylated at tyrosine residues following TGF-ß treatment, which initiated a signaling cascade resulting in the sequential recruitment of p85 subunit of PI3K and focal adhesion kinase (FAK). The SchA-FAK-p85 complex subsequently selectively recruited and activated Akt2, not Akt1. Inhibition of the p85 subunit through phosphorylated 1257 peptide completely attenuated EMT in these cells. We have defined the underlying mechanism responsible for isoform-specific recruitment and activation of Akt2, not Akt1, during TGF-ß-mediated EMT in A549 cells. Inhibition of the formation of this complex thus represents an important and novel therapeutic target in metastatic lung carcinoma.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Humanos , Fosforilação , Isoformas de Proteínas/metabolismo , Proteínas de Ligação a RNA
11.
PLoS One ; 9(4): e94132, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24718026

RESUMO

The phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is frequently activated in human cancer and plays a crucial role in glioblastoma biology. We were interested in gaining further insight into the potential of targeting PI3K isoforms as a novel anti-tumor approach in glioblastoma. Consistent expression of the PI3K catalytic isoform PI3K p110α was detected in a panel of glioblastoma patient samples. In contrast, PI3K p110ß expression was only rarely detected in glioblastoma patient samples. The expression of a module comprising the epidermal growth factor receptor (EGFR)/PI3K p110α/phosphorylated ribosomal S6 protein (p-S6) was correlated with shorter patient survival. Inhibition of PI3K p110α activity impaired the anchorage-dependent growth of glioblastoma cells and induced tumor regression in vivo. Inhibition of PI3K p110α or PI3K p110ß also led to impaired anchorage-independent growth, a decreased migratory capacity of glioblastoma cells, and reduced the activation of the Akt/mTOR pathway. These effects were selective, because targeting of PI3K p110δ did not result in a comparable impairment of glioblastoma tumorigenic properties. Together, our data reveal that drugs targeting PI3K p110α can reduce growth in a subset of glioblastoma tumors characterized by the expression of EGFR/PI3K p110α/p-S6.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/enzimologia , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Embrião de Galinha , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Ensaios de Seleção de Medicamentos Antitumorais , Indução Enzimática , Glioblastoma/enzimologia , Humanos , Hidrazonas/farmacologia , Morfolinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Sulfonamidas/farmacologia , Tiofenos/farmacologia , Células Tumorais Cultivadas
12.
Nucleic Acids Res ; 41(2): 855-68, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23175608

RESUMO

Genomic integrity is preserved by the action of protein complexes that control DNA homeostasis. These include the sliding clamps, trimeric protein rings that are arranged around DNA by clamp loaders. Replication factor C (RFC) is the clamp loader for proliferating cell nuclear antigen, which acts on DNA replication. Other processes that require mobile contact of proteins with DNA use alternative RFC complexes that exchange RFC1 for CTF18 or RAD17. Phosphoinositide 3-kinases (PI3K) are lipid kinases that generate 3-poly-phosphorylated-phosphoinositides at the plasma membrane following receptor stimulation. The two ubiquitous isoforms, PI3Kalpha and PI3Kbeta, have been extensively studied due to their involvement in cancer and nuclear PI3Kbeta has been found to regulate DNA replication and repair, processes controlled by molecular clamps. We studied here whether PI3Kbeta directly controls the process of molecular clamps loading. We show that PI3Kbeta associated with RFC1 and RFC1-like subunits. Only when in complex with PI3Kbeta, RFC1 bound to Ran GTPase and localized to the nucleus, suggesting that PI3Kbeta regulates RFC1 nuclear import. PI3Kbeta controlled not only RFC1- and RFC-RAD17 complexes, but also RFC-CTF18, in turn affecting CTF18-mediated chromatid cohesion. PI3Kbeta thus has a general function in genomic stability by controlling the localization and function of RFC complexes.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Proteína de Replicação C/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/química , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Reparo do DNA , Replicação do DNA , Humanos , Subunidades Proteicas/metabolismo , Proteína de Replicação C/antagonistas & inibidores , Proteína de Replicação C/química , Proteína ran de Ligação ao GTP/metabolismo
13.
J Pharmacol Sci ; 120(4): 270-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23149576

RESUMO

Studies with knockout mice have indicated that the only isoform of phosphoinositide 3-kinase (PI3K) functioning in the oxidative burst of mouse neutrophils in response to heterotrimeric guanine nucleotide-binding protein-coupled receptor (GPCR) agonists is a class-IB PI3K, p110γ. In the present study, we observed that the cells from p110γ(-/-) mice gain a response to N-formyl-Met-Leu-Phe (fMLP) after priming with cytochalasin E. Even the unprimed cells, which show no response to fMLP, produce a significant amount of superoxide, when an effective agonist of the mouse-type fMLP receptors, Trp-Lys-Tyr-Met-Val-D-Met, is used to stimulate the cells. These results suggested that the class-IA isoforms (p110α, p110ß, and p110δ) of PI3K are sufficient to trigger and maintain superoxide production. Examination of the effects of isoform-specific inhibitors suggested that the p110ß isoform is the primary PI3K triggering the response to GPCR agonists when p110γ is absent.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Neutrófilos/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Superóxidos/metabolismo , Animais , Células Cultivadas , Isoenzimas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/agonistas
14.
J Bone Miner Res ; 27(12): 2464-75, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22806988

RESUMO

Class IA phosphatidylinositol 3-kinases (PI3Ks) are activated by growth factor receptors and regulate a wide range of cellular processes. In osteoclasts, they are activated downstream of α(v) ß(3) integrin and colony-stimulating factor-1 receptor (c-Fms), which are involved in the regulation of bone-resorbing activity. The physiological relevance of the in vitro studies using PI3K inhibitors has been of limited value, because they inhibit all classes of PI3K. Here, we show that the osteoclast-specific deletion of the p85 genes encoding the regulatory subunit of the class IA PI3K results in an osteopetrotic phenotype caused by a defect in the bone-resorbing activity of osteoclasts. Class IA PI3K is required for the ruffled border formation and vesicular transport, but not for the formation of the sealing zone. p85α/ß doubly deficient osteoclasts had a defect in macrophage colony-stimulating factor (M-CSF)-induced protein kinase B (Akt) activation and the introduction of constitutively active Akt recovered the bone-resorbing activity. Thus, the class IA PI3K-Akt pathway regulates the cellular machinery crucial for osteoclastic bone resorption, and may provide a molecular basis for therapeutic strategies against bone diseases.


Assuntos
Reabsorção Óssea/fisiopatologia , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Animais , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Osteoclastos/fisiologia , Osteopetrose/patologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
15.
Blood ; 119(17): 3951-61, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-22378847

RESUMO

We show that loss of p85α inhibits the growth and maturation of mast cells, whereas loss of p85ß enhances this process. Whereas restoring the expression of p85α in P85α(-/-) cells restores these functions, overexpression of p85ß has the opposite effect. Consistently, overexpression of p85ß in WT mast cells represses KIT-induced proliferation and IL-3-mediated maturation by inhibiting the expression of Microphthalmia transcription factor. Because p85α and p85ß differ in their N-terminal sequences, chimeric proteins consisting of amino or carboxy-terminal of p85α and/or p85ß do not rescue the growth defects of p85α(-/-) cells, suggesting cooperation between these domains for normal mast cell function. Loss of p85ß impaired ligand induced KIT receptor internalization and its overexpression enhanced this process, partly because of increased binding of c-Cbl to p85ß relative to p85α. In vivo, loss of p85ß resulted in increased mast cells, and bone marrow transplantation of cells overexpressing p85ß resulted in significant reduction in some tissue mast cells. Overexpression of p85ß suppressed the growth of oncogenic KIT-expressing cells in vitro and prolonged the survival of leukemic mice in vivo. Thus, p85α and p85ß differentially regulate SCF and oncogenic KIT-induced signals in myeloid lineage-derived mast cells.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Leucemia/etiologia , Leucemia/patologia , Mastócitos/patologia , Animais , Western Blotting , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Leucemia/metabolismo , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real
16.
PLoS One ; 7(1): e28979, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22238586

RESUMO

Stem cell factor (SCF) mediated KIT receptor activation plays a pivotal role in mast cell growth, maturation and survival. However, the signaling events downstream from KIT are poorly understood. Mast cells express multiple regulatory subunits of class 1(A) PI3Kinase (PI3K) including p85α, p85ß, p50α, and p55α. While it is known that PI3K plays an essential role in mast cells; the precise mechanism by which these regulatory subunits impact specific mast cell functions including growth, survival and cycling are not known. We show that loss of p85α impairs the growth, survival and cycling of mast cell progenitors (MCp). To delineate the molecular mechanism (s) by which p85α regulates mast cell growth, survival and cycling, we performed microarray analyses to compare the gene expression profile of MCps derived from WT and p85α-deficient mice in response to SCF stimulation. We identified 151 unique genes exhibiting altered expression in p85α-deficient cells in response to SCF stimulation compared to WT cells. Functional categorization based on DAVID bioinformatics tool and Ingenuity Pathway Analysis (IPA) software relates the altered genes due to lack of p85α to transcription, cell cycle, cell survival, cell adhesion, cell differentiation, and signal transduction. Our results suggest that p85α is involved in mast cell development through regulation of expression of growth, survival and cell cycle related genes.


Assuntos
Diferenciação Celular/genética , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Genes cdc/fisiologia , Mastócitos/fisiologia , Animais , Ciclo Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Análise por Conglomerados , Mastócitos/metabolismo , Camundongos , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/classificação , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
17.
Int J Immunopathol Pharmacol ; 25(4): 967-76, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23298487

RESUMO

Bone morphogenetic protein 2 (BMP-2) is a member of the TGF-beta superfamily of signaling molecules, and has been shown to function as a tumor suppressor involved in development and progression of many malignancies. BMP-2 has previously been reported to be closely correlated with lung cancer. But, the role and molecular mechanisms of BMP-2 in lung cancer have not yet been comprehensively explained. The present study aims to elucidate the role of BMP-2 in growth and invasion of human lung adenocarcinoma (LAC) in vitro and in vivo. Adenovirus vector-mediated BMP-2 small hairpin RNA (shBMP-2) was used to transfect into A549 LAC cells to determine the functional relevance of BMP-2 and tumor growth and invasion in vitro and in vivo, and further investigate the expression levels of BMP-2, vascular endothelial growth factor (VEGF), matrix metallopeptidase-9 (MMP-9), phosphatidylinositol 3-kinase p85alpha (PI3Kp85alpha) and phosphorylated AKT (p-AKT). As a result, LAC cell proliferation and invasion were significantly diminished by knockdown of BMP-2 indicated by MTT and Transwell assays, and cell apoptosis and cycle arrest were markedly induced indicated by flow cytometry. When BMP-2 expression was knocked down, the expression of PI3Kp85alpha, p-AKT, VEGF and MMP-9 was also down-regulated in LAC cells. In addition, the tumor volumes in LAC subcutaneous nude mouse model treated with shBMP-2 were significantly smaller than those in control and ad-GFP groups. Taken together, our findings indicate that knockdown of BMP-2 inhibits growth and invasion of LAC cells possibly via blockade of the PI3K/AKT signaling pathway, and BMP-2 may be a potential therapeutic target for lung cancer.


Assuntos
Proteína Morfogenética Óssea 2/antagonistas & inibidores , Neoplasias Pulmonares/terapia , RNA Interferente Pequeno/genética , Adenoviridae/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/fisiologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Classe Ia de Fosfatidilinositol 3-Quinase/análise , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/patologia , Metaloproteinase 9 da Matriz/análise , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-akt/fisiologia , Fator A de Crescimento do Endotélio Vascular/análise
18.
Mol Cell Endocrinol ; 356(1-2): 24-30, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-21684319

RESUMO

Phosphatidylinositol 3-kinase (PI3K) signaling is a fundamental pathway for the regulation of cell proliferation, survival, migration, and metabolism in a variety of physiological and pathological processes. In recent years information provided by genetically modified mouse models has revealed that PI3K signaling plays vital roles in oogenesis, folliculogenesis, ovulation, and carcinogenesis in mouse ovary. In this review, we summarize (1) the physiological function of intra-oocyte PI3K signaling in regulation of primordial follicle survival and activation; (2) intra-granulosa cell PI3K signaling in regulation of cyclic follicular recruitment and ovulation; (3) intra-oocyte PI3K signaling in regulation of meiosis resumption and early embryogenesis; and also (4) the pathological function of PI3K signaling in ovarian diseases such as premature ovarian failure, granulosa cell tumors, and ovarian surface epithelium carcinomas. This updated info hopefully will lead to a better understanding of the human ovary and provide potential therapies for treating human infertility.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Ovário/fisiologia , Transdução de Sinais , Animais , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Meiose , Ovário/citologia , Ovário/embriologia , Ovário/enzimologia
19.
Eur J Cancer ; 48(1): 149-57, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22079609

RESUMO

Glioblastoma multiforme (GBM) is a highly invasive and aggressive primary brain tumour in which loss of phosphatase and tensin homologue deleted on chromosome 10 (PTEN), a negative regulator of PI3K signalling, is a common feature. PTEN/PI3K/Akt signalling is involved in the regulation of proliferation, apoptosis and cell migration. Deregulation of PI3K signalling is considered an essential driver in gliomagenesis. However, the role of different PI3K isoforms in glioma is still largely unclear. Here we show that the catalytic PI3K isoform p110δ is consistently expressed at a high level in various glioma cell lines. We used small interfering RNA to selectively deplete p110δ and to determine its tumourigenic roles in PTEN-deficient cells. Interestingly, knockdown of p110δ decreased the cell migration and invasion ability of all GBM cell lines tested. Mechanistically, p110δ knockdown reduced the protein levels of focal adhesion kinase and cell division cycle 42, key regulators of cellular migration. In contrast, pharmacologic inhibition of p110δ by IC87114 or CAL-101 also clearly impaired glioma cell migration but had no obvious effect on the invasion capacity thus pinpointing to possible kinase-dependent and -independent roles of p110δ in glioma pathology. In summary, our data provide novel evidence that in glioma cells p110δ is a key regulator of cell movement and thus may contribute to the highly invasive phenotype of GBM. Isoform specific targeting of PI3Kδ may be beneficial in the treatment of glioblastoma multiforme by specifically inhibiting tumour cell migration capacity.


Assuntos
Neoplasias Encefálicas/patologia , Movimento Celular/genética , Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Glioma/patologia , Neoplasias Encefálicas/genética , Domínio Catalítico/genética , Domínio Catalítico/fisiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glioma/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Invasividade Neoplásica , Inibidores de Fosfoinositídeo-3 Quinase , RNA Interferente Pequeno/farmacologia , Proteína cdc42 de Ligação ao GTP/metabolismo
20.
Cell Cycle ; 10(21): 3731-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22045127

RESUMO

Addition of short (6 to 16 amino acids) peptide sequences to the N-terminus of p110α induces a gain of function. Such sequences include the common Flag, His, and VSV tags as well as random sequences. An N-terminal myristylation signal generally believed to activate p110α by providing a constitutive membrane address is also activating, if myristylation is mutationally abolished. The gain of function seen with N-terminally tagged (NTT) p110α constructs extends to signaling, oncogenic transformation and stimulation of cell growth. The activating effect of N-terminal tags requires a functional Ras-binding domain in p110α. Mutations in that domain (T208D and K227A) abolish the gains of function in oncogenicity and signaling. The dominant negative mutant of Ras, RasN17, interferes with transformation induced by NTT p110α. In contrast, binding to p85 activity is not required for cellular transformation and enhanced signaling by NTT p110α.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Transdução de Sinais , Sequência de Aminoácidos , Animais , Domínio Catalítico , Transformação Celular Neoplásica , Embrião de Galinha , Classe Ia de Fosfatidilinositol 3-Quinase/química , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA