Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Biol Int ; 48(4): 440-449, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38115179

RESUMO

Kirsten rat sarcoma virus (KRAS) gene mutation is common in colorectal cancer (CRC) and is often predictive of treatment failure and poor prognosis. To understand the mechanism, we compared the transcriptome of CRC patients with wild-type and mutant KRAS and found that KRAS mutation is associated with the overexpression of a secreted serine protease, kallikrein-related peptidase 10 (KLK10). Moreover, using in vitro and in vivo models, we found that KLK10 overexpression favors the rapid growth and liver metastasis of KRAS mutant CRC and can also impair the efficacy of KRAS inhibitors, leading to drug resistance and poor survival. Further functional assays revealed that the oncogenic role of KLK10 is mediated by protease-activated receptor 1 (PAR1). KLK10 cleaves and activates PAR1, which further activates 3-phosphoinositide-dependent kinase 1 (PDK1)-AKT oncogenic pathway. Notably, suppressing PAR1-PDK1-AKT cascade via KLK10 knockdown can effectively inhibit CRC progression and improve the sensitivity to KRAS inhibitor, providing a promising therapeutic strategy. Taken together, our study showed that KLK10 promotes the progression of KRAS mutant CRC via activating PAR1-PDK1-AKT signaling pathway. These findings expanded our knowledge of CRC development, especially in the setting of KRAS mutation, and also provided novel targets for clinical intervention.


Assuntos
Neoplasias Colorretais , Receptor PAR-1 , Humanos , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Calicreínas/genética , Calicreínas/metabolismo , Mutação/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Transdução de Sinais , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo
2.
J Biol Chem ; 295(24): 8120-8134, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32350110

RESUMO

Protein kinase B (AKT1) is a central node in a signaling pathway that regulates cell survival. The diverse pathways regulated by AKT1 are communicated in the cell via the phosphorylation of perhaps more than 100 cellular substrates. AKT1 is itself activated by phosphorylation at Thr-308 and Ser-473. Despite the fact that these phosphorylation sites are biomarkers for cancers and tumor biology, their individual roles in shaping AKT1 substrate selectivity are unknown. We recently developed a method to produce AKT1 with programmed phosphorylation at either or both of its key regulatory sites. Here, we used both defined and randomized peptide libraries to map the substrate selectivity of site-specific, singly and doubly phosphorylated AKT1 variants. To globally quantitate AKT1 substrate preferences, we synthesized three AKT1 substrate peptide libraries: one based on 84 "known" substrates and two independent and larger oriented peptide array libraries (OPALs) of ∼1011 peptides each. We found that each phospho-form of AKT1 has common and distinct substrate requirements. Compared with pAKT1T308, the addition of Ser-473 phosphorylation increased AKT1 activities on some, but not all of its substrates. This is the first report that Ser-473 phosphorylation can positively or negatively regulate kinase activity in a substrate-dependent fashion. Bioinformatics analysis indicated that the OPAL-activity data effectively discriminate known AKT1 substrates from closely related kinase substrates. Our results also enabled predictions of novel AKT1 substrates that suggest new and expanded roles for AKT1 signaling in regulating cellular processes.


Assuntos
Proteínas Proto-Oncogênicas c-akt/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Humanos , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/metabolismo , Fosforilação , Fosfosserina/metabolismo , Proteínas Proto-Oncogênicas c-akt/química , Curva ROC , Especificidade por Substrato
3.
Reprod Domest Anim ; 55(12): 1678-1687, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32946622

RESUMO

Oocyte maturation plays a vitally important role in porcine reproduction. Regrettably, the quality of oocytes matured in vitro is weaker than that of in vivo matured oocytes. We collected and cultivated porcine cumulus oocyte complexes (COCs) in vitro with phosphoinositide-dependent kinase 1 (PDK1) activator 5-(4-chloro-phenyl)-3-phenyl-pent-2-enoic acid (PS48), whose concentrations were 0, 2, 5, 10 and 20 µM to investigate whether the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) signalling pathway would impact the oocyte quality. The results showed that 10 µM PS48 increased the oocyte proportion of metaphase II (MII) stage and improved the expansion of cumulus cells (CCs). What's more, the activation of PI3K/Akt signalling pathway could regulate the expression of maturation-related genes and proteins. The results of quantitative real-time PCR showed that 10 µM PS48 increased the mRNA and protein levels of Akt and regulated maturation-related genes, including cyclin B1, MOS, BMP15, GDF9, CDC2, mTOR, BAX, BCL2 and caspase-3. The results of Western blot indicated that 10µM PS48 increased the protein abundance of Akt, phosphorylation of Akt Thr308 (p-AktThr308 ) and cyclin B1, but decreased the protein abundance of pro-apoptotic BAX. These results suggested that adding 10 µM PS48 to mature culture medium could promote the maturation of porcine oocytes, potentially through activating the PI3K/Akt signalling pathway.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Técnicas de Maturação in Vitro de Oócitos/veterinária , Suínos/fisiologia , Animais , Células do Cúmulo/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Maturação in Vitro de Oócitos/métodos , Oócitos/efeitos dos fármacos , Oócitos/crescimento & desenvolvimento , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
4.
J Cell Biochem ; 120(1): 126-134, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30218446

RESUMO

Clear cell renal cell carcinoma (ccRCC) is the most popular kidney cancer in adults. Metabolic shift toward aerobic glycolysis is a fundamental factor for ccRCC therapy. MicroRNAs (miRNAs) are thought to be important regulators in ccRCC development and progression. Phosphoinositide-dependent kinase 1 (PDK1) is required for metabolic activation; however, the role of PDK1-induced glycolytic metabolism regulated by miRNAs is unclear in ccRCC. So, the purpose of the current study is to elucidate the underlying mechanism in ccRCC cell metabolism mediated by PDK1. Our results revealed that miR-409-3p inhibited glycolysis by regulating PDK1 expression in ccRCC cells. We also found that miR-409-3p was regulated by hypoxia. Our results indicated that PDK1 facilitated ccRCC cell glycolysis, regulated by miR-409-3p in hypoxia.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Carcinoma de Células Renais/metabolismo , Glicólise , Neoplasias Renais/metabolismo , MicroRNAs/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/genética , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/cirurgia , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/patologia , Neoplasias Renais/cirurgia , MicroRNAs/química , Mimetismo Molecular/genética , Consumo de Oxigênio , Transfecção
5.
Pflugers Arch ; 468(4): 667-77, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26577585

RESUMO

Hyperinsulinemia associated with type 2 diabetes may contribute to the development of vascular diseases. Although we recently reported that enhanced contractile responses to serotonin (5-hydroxytryptamine, 5-HT) are observed in the arteries of type 2 diabetes models, the causative factors and detailed signaling pathways involved remain unclear. The purpose of this study was to investigate whether high insulin would be an amplifier of 5-HT-induced contraction in rat carotid arteries and whether the contraction involves phosphoinositide 3-kinase (PI3K)/3-phosphoinositide-dependent protein kinase 1 (PDK1) signaling, an insulin-mediated signaling pathway. In rat carotid arteries organ-cultured with insulin (for 24 h), (1) the contractile responses to 5-HT were significantly greater (vs. vehicle), (2) the insulin-induced enhancement of 5-HT-induced contractions was largely suppressed by inhibitors of the insulin receptor (IR) (GSK1838705A), PI3K (LY294002), and PDK1 (GSK2334470), and (3) the levels of phosphorylated forms of both PDK1 and myosin phosphatase target subunit 1 (MYPT1) were greater upon 5-HT stimulation. In addition, in rat carotid arteries organ-cultured with an activator of PDK1 (PS48), the 5-HT-induced contraction was greater, and this was suppressed by PDK1 inhibition but not PI3K inhibition. In addition, MYPT1 and PDK1 phosphorylation upon 5-HT stimulation was enhanced (vs. vehicle). These results suggest that high insulin levels amplify 5-HT-induced contraction. Moreover, the present results indicated the direct linkage between IR/PI3K/PDK1 activation and 5-HT-induced contraction in rat carotid arteries for the first time.


Assuntos
Artérias Carótidas/metabolismo , Insulina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor de Insulina/metabolismo , Serotonina/metabolismo , Vasoconstrição , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/fisiologia , Masculino , Contração Muscular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Proteína Fosfatase 1/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Ratos , Ratos Wistar , Receptor de Insulina/antagonistas & inibidores , Transdução de Sinais
6.
Biochem Biophys Res Commun ; 470(1): 205-212, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26773505

RESUMO

The vital importance of the Leloir pathway of galactose metabolism has been repeatedly demonstrated by various uni-/multicellular model organisms, as well human patients who have inherited deficiencies of the key GAL enzymes. Yet, other than the obvious links to the glycolytic pathway and glycan biosynthetic pathways, little is known about how this metabolic pathway interacts with the rest of the metabolic and signaling networks. In this study, we compared the growth and the expression levels of the key components of the PI3K/Akt growth signaling pathway in primary fibroblasts derived from normal and galactose-1 phosphate uridylyltransferase (GalT)-deficient mice, the latter exhibited a subfertility phenotype in adult females and growth restriction in both sexes. The growth potential and the protein levels of the pAkt(Thr308), pAkt(Ser473), pan-Akt, pPdk1, and Hsp90 proteins were significantly reduced by 62.5%, 60.3%, 66%, 66%, and 50%, respectively in the GalT-deficient cells. Reduced expression of phosphorylated Akt proteins in the mutant cells led to diminished phosphorylation of Gsk-3ß (-74%). Protein expression of BiP and pPten were 276% and 176% higher respectively in cells with GalT-deficiency. Of the 24 genes interrogated using QIAGEN RT(2) Profiler PCR Custom Arrays, the mRNA abundance of Akt1, Pdpk1, Hsp90aa1 and Pi3kca genes were significantly reduced at least 2.03-, 1.37-, 2.45-, and 1.78-fold respectively in mutant fibroblasts. Both serum-fasted normal and GalT-deficient cells responded to Igf-1-induced activation of Akt phosphorylation at +15 min, but the mutant cells have lower phosphorylation levels. The steady-state protein abundance of Igf-1 receptor was also significantly reduced in mutant cells. Our results thus demonstrated that GalT deficiency can effect down-regulation of the PI3K/Akt growth signaling pathway in mouse fibroblasts through distinct mechanisms targeting both gene and protein expression levels.


Assuntos
Fibroblastos/metabolismo , Galactosemias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , UTP-Hexose-1-Fosfato Uridililtransferase/metabolismo , Animais , Estresse do Retículo Endoplasmático , Feminino , Galactosemias/patologia , Regulação Enzimológica da Expressão Gênica , Masculino , Camundongos
7.
Bioorg Med Chem ; 22(15): 3879-86, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25012568

RESUMO

PDK1, a biological target that has attracted a large amount of attention recently, is responsible for the positive regulation of the PI3K/Akt pathway that is often activated in a large number of human cancers. A series of second-generation 2-anilino-4-substituted-7H-pyrrolopyrimidines were synthesised by installation of various functions at the 4-position of the 7H-pyrrolopyrimidine scaffold. All compounds were screened against the isolated PDK1 enzyme and dose response analysis was obtained on the best compounds of the series.


Assuntos
Inibidores de Proteínas Quinases/síntese química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirimidinas/química , Pirróis/química , Humanos , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/síntese química , Pirimidinas/metabolismo , Pirróis/síntese química , Pirróis/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Relação Estrutura-Atividade
8.
Bioorg Med Chem ; 22(14): 3781-90, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24856302

RESUMO

PDK1 is an important regulator of the PI3K/Akt pathway, which has been found frequently activated in a large number of human cancers. Herein we described the preparation of novel substituted 3-anilino-quinolin-2(1H)-ones as PDK1 inhibitors. The synthesis is based around a Buchwald-Hartwig cross-coupling of various 3-bromo-6-substituted-quinolin-2(1H)-ones with three different functionalised anilines. The modular nature of the designed synthesis allowed access to a series of novel inhibitors through derivatisation of a late-stage intermediate. All compounds were screened against isolated PDK1 enzyme, with modest inhibition observed.


Assuntos
Compostos de Anilina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Compostos de Anilina/síntese química , Compostos de Anilina/química , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Quinolonas/síntese química , Quinolonas/química , Relação Estrutura-Atividade
9.
Recent Pat Anticancer Drug Discov ; 19(2): 253-255, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-36852816

RESUMO

The patent describes novel useful compounds, such as PI3K protein kinase inhibitors, in particular as PI3K delta (δ) and/or gamma (γ) protein kinase modulators. The present disclosure also provides methods for preparing PI3K protein kinase inhibitors, pharmaceutical compositions containing them, and methods of treatment, prevention, and amelioration of PI3K kinase-mediated diseases, and disorders.


Assuntos
Neoplasias , Fosfatidilinositol 3-Quinases , Humanos , Inibidores de Fosfoinositídeo-3 Quinase , Patentes como Assunto , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias/tratamento farmacológico , Fosfatidilinositol 3-Quinase
10.
Curr Diabetes Rev ; 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38468518

RESUMO

The role of 3-phosphoinositide-dependent kinase 1 (PDK1) has been welldocumented in the development of diabetes. This review offers a thorough examination of its composition and associated routes, specifically focusing on insulin signaling and glucose processing. By examining the precise connection between PDK1 and diabetes, various strategies specifically targeting PDK1 were also investigated. Additionally, recent discoveries from mouse models were compiled where PDK1 was knocked out in certain tissues, which demonstrated encouraging outcomes for focused treatments despite the absence of any currently approved clinical PDK1 activators. Moreover, the dual nature of PDK1 activation was discussed, encompassing both anti-diabetic and pro-oncogenic effects. Hence, the development of a PDK1 modifier is of utmost importance, as it can activate anti-diabetic pathways while inhibiting pro-oncogenic pathways, thus aiding in the treatment of diabetes. In general, PDK1 presents a noteworthy opportunity for future therapeutic strategies in the treatment of diabetes.

11.
J Cancer ; 15(5): 1429-1441, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38356720

RESUMO

Background: Renal cell carcinoma (RCC) frequently exhibits activating PI3K-Akt-mTOR pathway mutations. 3-Phosphoinositide-dependent kinase 1 (PDPK1 or PDK1) has been established to play a pivotal role in modulating PI3K pathway signaling. mTOR is the main autophagy-initiating factor. However, limited advances have been made in understanding the relationship between PDPK1 and autophagy in RCC. Methods: GSK2334470 (GSK470), a novel and highly specific inhibitor of PDPK1, was selected to investigate the anticancer effects in two RCC cell lines. Cell growth was assessed by CCK-8 test and colony formation. Changes in the protein levels of key Akt/mTOR pathway components and apoptosis markers were assessed by Western blotting. Autophagy was assessed by using LC3B expression, transmission electron microscopy, and a tandem mRFP-EGFP-LC3 construct. The effect of PDPK1 and autophagy inhibitor chloroquine in RCC in vivo was examined in a mouse tumor-bearing model. Results: GSK470 significantly inhibited cell proliferation and induces apoptosis in A498 and 786-O RCC cells. GSK470 downregulates the phosphorylation of PDPK1, thereby inhibiting downstream phosphorylation of Akt1 at Thr308 and Ser473 and mTOR complex 1 (mTORC1) activity. Treatment with insulin-like growth factor-1 (IGF-1) partially restored GSK470-induced behaviors/activities. Interestingly, treatment of A498 and 786-O cells with GSK470 or siPDPK1 induced significant increases in the hallmarks of autophagy, including autophagosome accumulation, autophagic flux, and LC3B expression. Importantly, GSK470 and chloroquine synergistically inhibited the growth of RCC cells in vitro and in xenograft models, supporting the protective role of autophagy activation upon blockade of the PDPK1-Akt-mTOR signaling pathway. Conclusion: Our study provides new insight into PDPK1 inhibition combined with autophagy inhibition as a useful treatment strategy for RCC.

12.
Diabetol Int ; 14(2): 119-124, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37090134

RESUMO

Adipose tissue regulates systemic energy metabolism through adipokine production as well as energy storage and energy supply to other organs in response to changes in energy status. Adipose tissue dysfunction is therefore thought to be a key contributor to the pathogenesis of a variety of metabolic disorders including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Given that insulin plays a central role in the regulation of many aspects of adipocyte function, insulin resistance in adipose tissue is implicated in the pathogenesis of metabolic disorders as a cause of adipose tissue dysfunction. The concept of metabolic dysfunction-associated fatty liver disease (MAFLD) has recently been proposed for liver disease associated with metabolic disorders in both obese and nonobese individuals, with insulin resistance in adipose tissue likely being an important factor in its pathogenesis. This review outlines the relation between insulin resistance in adipose tissue and metabolic disorders, with a focus on the physiological relevance and mechanism of action of 3'-phosphoinositide-dependent kinase 1 (PDK1), a key kinase in insulin signaling, and its downstream transcription factor FoxO1 in adipocytes.

13.
J Biomol Struct Dyn ; : 1-12, 2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37646644

RESUMO

PDK1, an attractive cancer target that downstreams 23 other kinases towards cell growth, survival and metabolism has gaining attention due to allosteric effect of ligands bound to it. Generally, the drug design strategy using pharmacophores is either a single protein structure or ensemble or ligand-based. Apart from these methods, yet another new approach of protein-protein docking with state of art computational tool like Schrodinger Suite to generate pharmacophores based on the interacting partners of the protein is proposed in this work. The structure-based pharmacophoric features were picked up from docking the ten interacting partners of PDK1 and screened against the Enamine libraries containing protein-protein interacting compound collection, advanced, protein mimetic and allosteric compounds. High throughput virtual screening against the PIF pocket of PDK1 yields an indole scaffold. The identified indole derivative is proposed to be a strong activator that binds in the protein-protein interaction site of PDK1 which was further confirmed by molecular metadynamics simulations, free energy surface analysis and MM-GBSA calculations. Thus, the pharmacophores generated by the interacting proteins for PPI can facilitate the virtual screening in structure-based drug discovery of similar therapeutic targets.Communicated by Ramaswamy H. Sarma.

14.
Cells ; 11(11)2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35681431

RESUMO

Alzheimer's disease (AD) is a common age-related neurodegenerative disease that leads to memory loss and cognitive function damage due to intracerebral neurofibrillary tangles (NFTs) and amyloid-ß (Aß) protein deposition. The phosphoinositide-dependent protein kinase (PDK1)/protein kinase B (Akt) signaling pathway plays a significant role in neuronal differentiation, synaptic plasticity, neuronal survival, and neurotransmission via the axon-dendrite axis. The phosphorylation of PDK1 and Akt rises in the brain, resulting in phosphorylation of the TNF-α-converting enzyme (TACE) at its cytoplasmic tail (the C-terminal end), changing its internalization as well as its trafficking. The current review aimed to explain the mechanisms of the PDK1/Akt/TACE signaling axis that exerts its modulatory effect on AD physiopathology. We provide an overview of the neuropathological features, genetics, Aß aggregation, Tau protein hyperphosphorylation, neuroinflammation, and aging in the AD brain. Additionally, we summarized the phosphoinositide 3-kinase (PI3K)/PDK1/Akt pathway-related features and its molecular mechanism that is dependent on TACE in the pathogenesis of AD. This study reviewed the relationship between the PDK1/Akt signaling pathway and AD, and discussed the role of PDK1/Akt in resisting neuronal toxicity by suppressing TACE expression in the cell membrane. This work also provides a perspective for developing new therapeutics targeting PDK1/Akt and TACE for the treatment of AD.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
15.
J Int Med Res ; 49(5): 3000605211016562, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34038205

RESUMO

BACKGROUND: Small-cell lung cancer (SCLC) accounts for approximately 15% to 20% of all lung cancers, and it is the leading cause of tumor-related deaths globally. This study explored the molecular mechanisms underlying the development of SCLC. METHODS: The correlations of phosphoinositide-dependent kinase-1 (PDPK1), p-Akt, and Hedgehog expression with patient characteristics were analyzed using SCLC specimens, and their expression was measured in BEAS-2B cells (control) and the SCLC cell lines H82, H69, H446, H146, and H526. Transfection experiments were performed to inhibit or activate gene expression in cells. We then measured the proliferation and migration of H146 cells. RESULTS: PDPK1, p-Akt, and Hedgehog expression was significantly higher in SCLC tissues, and their expression was correlated with patient characteristics. p-Akt expression was significantly correlated with Hedgehog expression. In H146 cells, PDPK1 and p-Akt were significantly upregulated. Silencing of PDPK1 or Akt and inhibition of Hedgehog significantly inhibited the proliferation and migration of H146 cells. PDPK1 and Akt affected Hedgehog expression, but Hedgehog did not affect PDPK1 or p-Akt expression. CONCLUSIONS: The interaction between the PDPK1-Akt pathway and the Hedgehog pathway influences the prognosis, growth, and migration of SCLC.


Assuntos
Proteínas Hedgehog , Neoplasias Pulmonares , 1-Fosfatidilinositol 4-Quinase , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Hedgehog/genética , Humanos , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas c-akt/genética
16.
Ann Transl Med ; 9(22): 1671, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34988180

RESUMO

BACKGROUND: Acute lung injury (ALI) is a common inflammatory disease of the lung. This study aimed to investigate the effect of 3-phosphoinositide-dependent kinase 1 (PDK1) interference on the levels of fibrosis and proinflammatory factors in lipopolysaccharide (LPS)-induced ALI and discuss the relevant mechanism. METHODS: An ALI model was established by intravenous injection of LPS treatment. A total of 24 Sprague-Dawley (SD) rats were randomly divided into 4 groups: sham group; ALI group; ALI + shRNA-NC group; and ALI + PDK1-shRNA group. Lung injury score, minute ventilation, lung volume, and airway resistance were used to evaluate lung function injury. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect PDK1 messenger RNA (mRNA) level. Western blot was performed to detect expression levels of PDK1, transforming growth factor-ß (TGF-ß), α-smooth muscle actin (α-SMA), toll-like receptor 4 (TLR4), p65, and myeloid differentiation primary response gene 88 (MyD88). The contents of interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) were detected by enzyme-linked immunosorbent assay (ELISA). The pathological changes and fibrosis of lung tissues were estimated by hematoxylin and eosin (H&E) and Masson staining. RESULTS: The results revealed that high lung injury score, low minute ventilation, low lung volume, and small airway resistance were present in the ALI group. Likewise, severe histopathological damage and fibrosis were apparent in the ALI group. Otherwise, contents of TNF-α, iNOS, IL-6, MCP-1, and levels of α-SMA, TGF-ß, TLR4, phosphorylated (p)-p65, and MyD88 were enhanced in the ALI group. Interestingly, pathological changes and fibrosis were improved significantly in the ALI + PDK1-shRNA group. Besides, knockdown of PDK1 reduced lung injury score and enhanced minute ventilation, lung volume, and airway resistance. Moreover, knockdown of PDK1 decreased the contents of TNF-α, iNOS, IL-6, MCP-1, and levels of TGF-ß, α-SMA, TLR4, p-p65, and MyD88. CONCLUSIONS: Knockdown of PDK1 protects LPS-induced ALI via attenuating activation of the nuclear factor-κB (NF-κB)/p65 pathway.

17.
Clin Transl Oncol ; 21(10): 1312-1318, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30759304

RESUMO

PURPOSE: Phosphoinositide-dependent kinase 1 (PDK1) is highly expressed in many solid tumors. And several studies have demonstrated that PDK1 has been an emerging and promising target for anti-cancer therapies. However, the role of PDK1 has not been studied so far in malignant pheochromocytoma (PCC). METHODS: In this study, immunohistochemical staining was performed to investigate the protein level of PDK1 in 63 PCC tissue samples, of which 49 were benign and 14 were malignant. In addition, we evaluated the effect of inhibition of PDK1 with siRNA on cell growth, apoptosis and invasive capacity in PC12 cells and identified the underlying mechanisms. RESULTS: We found that PDK1 was overexpressed in malignant PCC tissues, and knockdown of PDK1 with siRNA significantly inhibited cell proliferation, increased apoptosis induction, and attenuated cell migration and invasive capacity in PC12 cells. We also showed that knockdown of PDK1 significantly reduced the phosphorylation of Akt at threonine 308 (p-Akt T308) but did not alter the serine phosphorylation of Akt on the S473 site (p-Akt S473). Furthermore, we found that the p-Akt expression was noticeably decreased after knockdown of PDK1, but the t-Akt expression did not show a significant decrease. CONCLUSION: We have demonstrated for the first time that PDK1 is overexpressed in human malignant PCC and plays an important role in the malignant biological behaviors of PC12 cell. Specifically, we have revealed that knockdown of PDK1 could attenuate activation of the Akt signaling. These data suggest that PDK1 could be a new promising potential therapeutic target in human cancer treatment for malignant PCC.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/análise , Neoplasias das Glândulas Suprarrenais/enzimologia , Proteínas de Neoplasias/análise , Feocromocitoma/enzimologia , Glândulas Suprarrenais/química , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno , Transfecção
18.
Expert Opin Ther Pat ; 29(4): 271-282, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30897008

RESUMO

INTRODUCTION: 3-Phosphoinositide-dependent kinase 1 (PDK1), the 'master kinase of the AGC protein kinase family', plays a key role in cancer development and progression. Although it has been rather overlooked, in the last decades a growing number of molecules have been developed to effectively modulate the PDK1 enzyme. AREAS COVERED: This review collects different PDK1 inhibitors patented from October 2014 to December 2018. The molecules have been classified on the basis of the chemical structure/type of inhibition, and for each general structure, examples have been discussed in extenso. EXPERT OPINION: The role of PDK1 in cancer development and progression as well as in metastasis formation and in chemoresistance has been confirmed by many studies. Therefore, the pharmaceutical discovery in both public and private institutions is still ongoing despite the plentiful molecules already published. The majority of the new molecules synthetized interact with binding sites different from the ATP binding site (i.e. PIF pocket or DFG-out conformation). However, many researchers are still looking for innovative PDK1 modulation strategy such as combination of well-known inhibitory agents or multitarget ligands, aiming to block, together with PDK1, other different critical players in the wide panorama of proteins involved in tumor pathways.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Trifosfato de Adenosina/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação , Progressão da Doença , Desenvolvimento de Medicamentos/métodos , Humanos , Ligantes , Neoplasias/enzimologia , Neoplasias/patologia , Patentes como Assunto
19.
Acta Pharm Sin B ; 8(5): 767-771, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30245963

RESUMO

The serum and glucocorticoid inducible protein kinase (SGK) family members share similar structure, substrate specificity and function with AKT and signal downstream of the phosphatidylinositol 3-kinase (PI3K) signalling pathway. They regulate a range of fundamental cellular processes such as cell proliferation and survival, thereby playing an important role in cancer development. This perspective intends to give an overview on the involvement of SGKs (particularly SGK3) in cancer progression, and compares the actions of SGK3 and AKT in cell cycle regulation, oncogenic signalling, and the potential as a therapeutic target for cancer.

20.
J Thromb Haemost ; 16(6): 1211-1225, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29575487

RESUMO

Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY: Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3ß. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Plaquetas/enzimologia , Proteínas Quinases Ativadas por Mitógeno/sangue , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/sangue , Embolia Pulmonar/enzimologia , Trombose/enzimologia , Tromboxanos/sangue , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/antagonistas & inibidores , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/sangue , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/deficiência , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/genética , Animais , Plaquetas/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Camundongos Knockout , Fosforilação , Inibidores da Agregação Plaquetária/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proto-Oncogene Mas , Embolia Pulmonar/sangue , Embolia Pulmonar/genética , Embolia Pulmonar/prevenção & controle , Pirimidinas/farmacologia , Transdução de Sinais , Tiofenos/farmacologia , Trombose/sangue , Trombose/genética , Trombose/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA