Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 71(2): 201-215.e7, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-30029001

RESUMEN

Macrophages are a dominant leukocyte population in the tumor microenvironment and actively promote cancer progression. However, the molecular mechanism underlying the role of macrophages remains poorly understood. Here we show that polarized M2 macrophages enhance 3-phosphoinositide-dependent protein kinase 1 (PDPK1)-mediated phosphoglycerate kinase 1 (PGK1) threonine (T) 243 phosphorylation in tumor cells by secreting interleukin-6 (IL-6). This phosphorylation facilitates a PGK1-catalyzed reaction toward glycolysis by altering substrate affinity. Inhibition of PGK1 T243 phosphorylation or PDPK1 in tumor cells or neutralization of macrophage-derived IL-6 abrogates macrophage-promoted glycolysis, proliferation, and tumorigenesis. In addition, PGK1 T243 phosphorylation correlates with PDPK1 activation, IL-6 expression, and macrophage infiltration in human glioblastoma multiforme (GBM). Moreover, PGK1 T243 phosphorylation also correlates with malignance and prognosis of human GBM. Our findings demonstrate a novel mechanism of macrophage-promoted tumor growth by regulating tumor cell metabolism, implicating the therapeutic potential to disrupt the connection between macrophages and tumor cells by inhibiting PGK1 phosphorylation.


Asunto(s)
Macrófagos/metabolismo , Fosfoglicerato Quinasa/genética , Fosfoglicerato Quinasa/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Glucólisis , Humanos , Macrófagos/patología , Ratones , Ratones Desnudos , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Fosforilación , Pronóstico , Microambiente Tumoral
2.
BMC Genomics ; 25(1): 360, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38605297

RESUMEN

BACKGROUND: During mitosis the cell depends on proper attachment and segregation of replicated chromosomes to generate two identical progeny. In cancers defined by overexpression or dysregulation of the MYC oncogene this process becomes impaired, leading to genomic instability and tumor evolution. Recently it was discovered that the chromatin regulator WDR5-a critical MYC cofactor-regulates expression of genes needed in mitosis through a direct interaction with the master kinase PDPK1. However, whether PDPK1 and WDR5 contribute to similar mitotic gene regulation in MYC-overexpressing cancers remains unclear. Therefore, to characterize the influence of WDR5 and PDPK1 on mitotic gene expression in cells with high MYC levels, we performed a comparative transcriptomic analysis in neuroblastoma cell lines defined by MYCN-amplification, which results in high cellular levels of the N-MYC protein. RESULTS: Using RNA-seq analysis, we identify the genes regulated by N-MYC and PDPK1 in multiple engineered CHP-134 neuroblastoma cell lines and compare them to previously published gene expression data collected in CHP-134 cells following inhibition of WDR5. We find that as expected N-MYC regulates a multitude of genes, including those related to mitosis, but that PDPK1 regulates specific sets of genes involved in development, signaling, and mitosis. Analysis of N-MYC- and PDPK1-regulated genes reveals a small group of commonly controlled genes associated with spindle pole formation and chromosome segregation, which overlap with genes that are also regulated by WDR5. We also find that N-MYC physically interacts with PDPK1 through the WDR5-PDPK1 interaction suggesting regulation of mitotic gene expression may be achieved through a N-MYC-WDR5-PDPK1 nexus. CONCLUSIONS: Overall, we identify a small group of genes highly enriched within functional gene categories related to mitotic processes that are commonly regulated by N-MYC, WDR5, and PDPK1 and suggest that a tripartite interaction between the three regulators may be responsible for setting the level of mitotic gene regulation in N-MYC amplified cell lines. This study provides a foundation for future studies to determine the exact mechanism by which N-MYC, WDR5, and PDPK1 converge on cell cycle related processes.


Asunto(s)
Genes myc , Neuroblastoma , Humanos , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Línea Celular Tumoral , Segregación Cromosómica , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Neuroblastoma/metabolismo
3.
J Gene Med ; 26(1): e3658, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38282149

RESUMEN

BACKGROUND: Aberrant activation of the phosphatidlinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway has been shown to play an important role in lung adenocarcinoma (LUAD). The effect of KRAS mutations, one of the important signatures of LUAD, on the PI3K/AKT/mTOR pathway in LUAD remains unclear. METHODS: The Seurat package and principal component analysis were used for cell categorization of single-cell RNA sequencing data of LUAD. The AUCell score was used to assess the activity of the PI3K/AKT/mTOR pathway. Meanwhile, using the gene expression profiles and mutation profiles in the The Cancer Genome Atlas dataset, LUAD patients were categorized into KRAS-mutant (KRAS-MT) and KRAS-wild-types (KRAS-WT), and the corresponding enrichment scores were calculated using gene set enrichment analysis analysis. Finally, the subpopulation of cells with the highest pathway activity was identified, the copy number variation profile of this subpopulation was inscribed using the inferCNV package and the CMap database was utilized to make predictions for drugs targeting this subpopulation. RESULTS: There is higher PI3K/AKT/mTOR pathway activity in LUAD epithelial cells with KRAS mutations, and high expression of KRAS, PIK3CA, AKT1 and PDPK1. In particular, we found significantly higher levels of pathway activity and associated gene expression in KRAS-MT than in KRAS-WT. We identified the highest pathway activity on a subpopulation of GRB2+ epithelial cells and the presence of amplified genes within its pathway. Finally, drugs were able to target GRB2+ epithelial cell subpopulations, such as wortmannin, palbociclib and angiogenesis inhibitor. CONCLUSIONS: The present study provides a basic theory for the activation of the PI3K/AKT/mTOR signaling pathway as a result of KRAS mutations.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Adenocarcinoma del Pulmón/genética , Variaciones en el Número de Copia de ADN , Neoplasias Pulmonares/patología , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Análisis de Secuencia de ARN , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
4.
Biochem Biophys Res Commun ; 721: 150106, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38795634

RESUMEN

3-phosphoinositide-dependent protein kinase 1 (PDK1) exhibits a substantial influence on immune cell development by establishing a vital connection between PI3K and downstream mTOR signaling cascades. However, it remains unclear whether PDK1 signaling affects the homeostasis and functionality of immune cells. To explore the impact of PDK1 on different immune cells within immune organs, transgenic mouse strains with lymphocyte-specific PDK1 knockout (PDK1fl/fl CD2-Cre) were generated. Unlike wild-type (WT) mice, lymphocyte-specific PDK1 knockout (KO) mice exhibited thymic atrophy, elevated percentages of CD8+ T cells and neutrophils, and reduced proportions of γδ T cells, B cells, and NK cells in the spleen. Functional analysis revealed elevated release of IFN-γ and IL-17A by T cells in PDK1 KO mice, contrasting with diminished levels observed in γδ T cells and Treg cells. Furthermore, the activation, cytotoxicity, and migratory potential of γδ T cells in PDK1 KO mice are heightened, indicating a potential association with the regulation of the mTOR signaling pathway. To conclude, the findings of this research demonstrated that specific knockout of PDK1 in lymphocytes hindered T cell development in the thymus and exhibited a substantial influence on immune cell homeostasis in the spleen and lymph nodes.


Asunto(s)
Ratones Noqueados , Timo , Animales , Ratones , Timo/inmunología , Bazo/inmunología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Transducción de Señal , Ratones Endogámicos C57BL , Serina-Treonina Quinasas TOR/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Interleucina-17/metabolismo , Interleucina-17/inmunología , Linfocitos T CD8-positivos/inmunología
5.
PLoS Biol ; 19(12): e3001483, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34879056

RESUMEN

Cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signalling is essential for the proliferation of Plasmodium falciparum malaria blood stage parasites. The mechanisms regulating the activity of the catalytic subunit PfPKAc, however, are only partially understood, and PfPKAc function has not been investigated in gametocytes, the sexual blood stage forms that are essential for malaria transmission. By studying a conditional PfPKAc knockdown (cKD) mutant, we confirm the essential role for PfPKAc in erythrocyte invasion by merozoites and show that PfPKAc is involved in regulating gametocyte deformability. We furthermore demonstrate that overexpression of PfPKAc is lethal and kills parasites at the early phase of schizogony. Strikingly, whole genome sequencing (WGS) of parasite mutants selected to tolerate increased PfPKAc expression levels identified missense mutations exclusively in the gene encoding the parasite orthologue of 3-phosphoinositide-dependent protein kinase-1 (PfPDK1). Using targeted mutagenesis, we demonstrate that PfPDK1 is required to activate PfPKAc and that T189 in the PfPKAc activation loop is the crucial target residue in this process. In summary, our results corroborate the importance of tight regulation of PfPKA signalling for parasite survival and imply that PfPDK1 acts as a crucial upstream regulator in this pathway and potential new drug target.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Dominio Catalítico , Línea Celular , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Eritrocitos/parasitología , Humanos , Malaria , Malaria Falciparum/parasitología , Merozoítos , Parásitos/metabolismo , Proteínas Protozoarias/metabolismo
6.
J Immunol ; 206(8): 1776-1783, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33789982

RESUMEN

Regulatory T (Treg) cells have an essential role in maintaining immune homeostasis, in part by suppressing effector T cell functions. Phosphoinositide-dependent kinase 1 (PDK1) is a pleiotropic kinase that acts as a key effector downstream of PI3K in many cell types. In T cells, PDK1 has been shown to be critical for activation of NF-κB and AKT signaling upon TCR ligation and is therefore essential for effector T cell activation, proliferation, and cytokine production. Using Treg cell-specific conditional deletion, we now demonstrate that PDK1 is also essential for Treg cell suppressive activity in vivo. Ablation of Pdk1 specifically in Treg cells led to systemic, lethal, scurfy-like inflammation in mice. Genome-wide analysis confirmed that PDK1 is essential for the regulation of key Treg cell signature gene expression and, further, suggested that PDK1 acts primarily to control Treg cell gene expression through regulation of the canonical NF-κB pathway. Consistent with these results, the scurfy-like phenotype of mice lacking PDK1 in Treg cells was rescued by enforced activation of NF-κB downstream of PDK1. Therefore, PDK1-mediated activation of the NF-κB signaling pathway is essential for regulation of Treg cell signature gene expression and suppressor function.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Trastornos Linfoproliferativos/genética , Linfocitos T Reguladores/inmunología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Antígenos CD4/metabolismo , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Terapia de Inmunosupresión , Activación de Linfocitos , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Transducción de Señal , Transcriptoma
7.
Proc Natl Acad Sci U S A ; 117(21): 11674-11684, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32393635

RESUMEN

Although adipocytes are major targets of insulin, the influence of impaired insulin action in adipocytes on metabolic homeostasis remains unclear. We here show that adipocyte-specific PDK1 (3'-phosphoinositide-dependent kinase 1)-deficient (A-PDK1KO) mice manifest impaired metabolic actions of insulin in adipose tissue and reduction of adipose tissue mass. A-PDK1KO mice developed insulin resistance, glucose intolerance, and hepatic steatosis, and this phenotype was suppressed by additional ablation of FoxO1 specifically in adipocytes (A-PDK1/FoxO1KO mice) without an effect on adipose tissue mass. Neither circulating levels of adiponectin and leptin nor inflammatory markers in adipose tissue differed between A-PDK1KO and A-PDK1/FoxO1KO mice. Lipidomics and microarray analyses revealed that leukotriene B4 (LTB4) levels in plasma and in adipose tissue as well as the expression of 5-lipoxygenase (5-LO) in adipose tissue were increased and restored in A-PDK1KO mice and A-PDK1/FoxO1KO mice, respectively. Genetic deletion of the LTB4 receptor BLT1 as well as pharmacological intervention to 5-LO or BLT1 ameliorated insulin resistance in A-PDK1KO mice. Furthermore, insulin was found to inhibit LTB4 production through down-regulation of 5-LO expression via the PDK1-FoxO1 pathway in isolated adipocytes. Our results indicate that insulin signaling in adipocytes negatively regulates the production of LTB4 via the PDK1-FoxO1 pathway and thereby maintains systemic insulin sensitivity.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Adipocitos/metabolismo , Araquidonato 5-Lipooxigenasa/metabolismo , Proteína Forkhead Box O1 , Resistencia a la Insulina , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Animales , Células Cultivadas , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Leucotrieno B4/metabolismo , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética
8.
Bull Exp Biol Med ; 174(4): 489-496, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36899199

RESUMEN

We explored the mechanism by which miR-139 modulates radioresistance of esophageal cancer (EC). The radioresistant cell line KYSE150R was obtained from the parental KYSE150 cell line by fractionated irradiation (15×2 Gy; total dose of 30 Gy). The cell cycle was assessed by flow cytometry. A gene profiling study was conducted to detect the expression of genes related to the radioresistance of EC. In the KYSE150R line, flow cytometry revealed increased number of G1-phase cells and decreased number of G2-phase cells; the expression of miR-139 increased. Knockdown of miR-139 decreased radioresistance and changed the distribution of cell cycle phases in KYSE150R cells. Western blotting showed that miR-139 knockdown increased the expression levels of cyclin D1, p-AKT, and PDK1. However, PDK1 inhibitor GSK2334470 reversed this effect for p-AKT and cyclin D1 expression. A luciferase reporter assay indicated that miR-139 directly bound to the PDK1 mRNA 3'-UTR. Analysis of the clinical data from 110 patients with EC showed an association of miR-139 expression with the TNM stage and the effect of therapy. MiR-139 expression significantly correlated with EC and progression-free survival. In conclusion, miR-139 enhances the radiosensitivity of EC by regulating the cell cycle through the PDK1/Akt/Cyclin D1 signaling pathway.


Asunto(s)
Neoplasias Esofágicas , MicroARNs , Tolerancia a Radiación , Humanos , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Línea Celular Tumoral/efectos de la radiación , Proliferación Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/radioterapia , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tolerancia a Radiación/genética , Transducción de Señal/genética
9.
PLoS Biol ; 17(10): e3000509, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31613895

RESUMEN

The Hippo signalling pathway restricts cell proliferation in animal tissues by inhibiting Yes-associated protein (YAP or YAP1) and Transcriptional Activator with a PDZ domain (TAZ or WW-domain-containing transcriptional activator [WWTR1]), coactivators of the Scalloped (Sd or TEAD) DNA-binding transcription factor. Drosophila has a single YAP/TAZ homolog named Yorkie (Yki) that is regulated by Hippo pathway signalling in response to epithelial polarity and tissue mechanics during development. Here, we show that Yki translocates to the nucleus to drive Sd-mediated cell proliferation in the ovarian follicle cell epithelium in response to mechanical stretching caused by the growth of the germline. Importantly, mechanically induced Yki nuclear localisation also requires nutritionally induced insulin/insulin-like growth factor 1 (IGF-1) signalling (IIS) via phosphatidyl inositol-3-kinase (PI3K), phosphoinositide-dependent kinase 1 (PDK1 or PDPK1), and protein kinase B (Akt or PKB) in the follicular epithelium. We find similar results in the developing Drosophila wing, where Yki becomes nuclear in the mechanically stretched cells of the wing pouch during larval feeding, which induces IIS, but translocates to the cytoplasm upon cessation of feeding in the third instar stage. Inactivating Akt prevents nuclear Yki localisation in the wing disc, while ectopic activation of the insulin receptor, PI3K, or Akt/PKB is sufficient to maintain nuclear Yki in mechanically stimulated cells of the wing pouch even after feeding ceases. Finally, IIS also promotes YAP nuclear localisation in response to mechanical cues in mammalian skin epithelia. Thus, the Hippo pathway has a physiological function as an integrator of epithelial cell polarity, tissue mechanics, and nutritional cues to control cell proliferation and tissue growth in both Drosophila and mammals.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasa/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transactivadores/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Animales , Fenómenos Biomecánicos , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Polaridad Celular , Proliferación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Células Epiteliales/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Larva/citología , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Mecanotransducción Celular , Ratones , Proteínas Nucleares/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transactivadores/metabolismo , Alas de Animales/citología , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo , Proteínas Señalizadoras YAP
10.
Zygote ; 30(3): 352-357, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34727997

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex disorder and genetic factors are believed to play a role. The main aim was to investigate expression levels of genes involved in PI3K/AKT signalling pathway pretreatment and post-treatment. Mouse models of PCOS were generated. Group one included control mice with no polycystic ovaries (n = 4), Group 2 included a PCOS mouse model (n = 8), Group 3 included PCOS mice treated with clomiphene citrate (n = 7) and Group 4 included PCOS mice treated with clomiphene citrate, metformin and pioglitazone (n = 8). Histochemical analyses were performed. Total RNA was extracted and cDNA was synthesized. Irs, Akt1 and Akt2, mTor and Pdpk1 gene expression levels were evaluated by RT-PCR amplification. In Group 1, cortex and medulla were evaluated as normal; in Group 2, ovarian cortex was composed of immature oocytes and cystic follicles with atretic follicles. In Groups 3 and 4, follicles were in the process of normal follicle differentiation. The expression levels of Akt1 and Pi3k were significantly different (P < 0.0001) between Groups 1 and 2. The significant differences in expression levels of Pi3k and Akt1 were also observed between the Group 1 and both Groups 3 and 4 (P < 0.0001). Furthermore, significant variations of the expression levels of mTor between Groups 1 and 4 were observed. The extrapolation of results of this study may imply that follicular development may be regulated by molecular pathways involving Pi3k, Akt1 and mTor expression. Therefore, genes in the PI3K/AKT pathway may have a direct regulatory role in the development of PCOS.


Asunto(s)
Síndrome del Ovario Poliquístico , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Clomifeno/farmacología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
11.
J Cell Physiol ; 236(7): 5432-5445, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33377210

RESUMEN

Osteoblasts are the main functional cells of bone formation, and they are responsible for the synthesis, secretion, and mineralization of the bone matrix. Phosphatidylinositol-3-kinase/Akt is an important signaling pathway involved in the regulation of cell proliferation, death, and survival. Some studies have shown that 3-phosphoinositide-dependent protein kinase-1 (PDK-1) plays an important role in the phosphorylation of Akt. In the present study, an osteocalcin (OCN) promoter-driven Cre-LoxP system was established to specifically delete the PDK-1 gene in osteoblasts. It was found that the size and weight of PDK-1 conditional gene knockout (cKO) mice were significantly reduced. von Kossa staining and microcomputed tomography showed that the trabecular thickness, trabecular number, and bone volume were significantly decreased, whereas trabecular separation was increased, as compared with wide-type littermates, which were characterized by a decreased bone mass. A model of distal femoral defect was established, and it was found that cKO mice delayed bone defect repair. In osteoblasts derived from PDK-1 cKO mice, the alkaline phosphatase (ALP) secretion and ability of calcium mineralization were significantly decreased, and the expressions of osteoblast-related proteins, runt-related transcription factor 2, OCN, and ALP were also clearly decreased. Moreover, the phosphorylation level of Akt and downstream factor GSK3ß and their response to insulin-like growth factor-1 (IGF-1) decreased clearly. Therefore, we believe that PDK-1 plays a very important role in osteoblast differentiation and bone formation by regulating the PDK-1/Akt/GSK3ß signaling pathway.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Regeneración Ósea/genética , Osteoblastos/metabolismo , Osteogénesis/genética , Animales , Diferenciación Celular/genética , Ratones , Ratones Noqueados
12.
Lab Invest ; 101(2): 165-176, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33199822

RESUMEN

Acute myocardial infarction (AMI) is a common clinical cardiovascular disease, which is the leading cause of death and disability worldwide. Abnormal expression of long noncoding RNAs (lncRNA) is reported to be related to myocardial dysfunctions such as myocardial infarction (MI). In this study, we aimed to investigate the role of lncRNA myocardial infarction-related transcription factors 2 (Mirt2) in AMI and the underlying molecular mechanisms in vivo and in vitro. In vivo AMI model was established by occlusion of the left anterior descending coronary artery. Rats were randomly divided into two groups (five rats per group): the sham group and the AMI group. H9c2 cells were cultured under hypoxia for 4 h and then cultured under normoxia to establish the in vitro hypoxia reoxygenation (H/R) model. Our study shows that the myocardial infarct size and the apoptosis in AMI rats were both significantly increased, indicating that the AMI rat model was successfully established. Additionally, the levels of Mirt2 in AMI rats were increased significantly. Knockdown of Mirt2 by shRNA (shMirt2) had no significant effect on apoptosis and MI in sham rats, but significantly promoted apoptosis and MI in AMI rats. In vitro experiments showed that shMirt2 significantly decreased the level of Mirt2 in H9c2 cells and H9c2 cells treated with H/R. It is worth noting that shMirt2 had no significant effect on H9c2 cells, but significantly increased the levels of oxidative stress markers (malondialdehyde and lactate dehydrogenase), and also increased the number of apoptosis of H/R-treated H9c2 cells. Further mechanistic analysis showed that Mirt2 could protect MI and apoptosis in AMI rats by competitively adsorbing miR-764 and reducing the inhibitory effect of miR-764 on 3-phosphoinositide-dependent kinase 1 (PDK1). More importantly, after overexpression of Mirt2, MI and apoptosis were significantly improved in AMI rats, indicating that Mirt2 showed a protective effect in AMI rats. In summary, these findings suggest that that Mirt2 participated in the regulation of MI through the miR-764/PDK1 axis. Therefore, the current findings provide a theoretical basis for the diagnosis and treatment of clinical MI with changes in Mirt2 levels.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Apoptosis/genética , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , ARN Largo no Codificante/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Línea Celular , Masculino , MicroARNs/genética , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley
13.
Exp Cell Res ; 386(2): 111746, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31778670

RESUMEN

Hypopharyngeal squamous cell carcinoma (HSCC) is a relatively rare malignancy and has the worst prognosis among head and neck cancer. Metastasis is the major cause of poor prognosis in HSCC patients. In this study, we found that 3-phosphoinositide-dependent protein kinase 1 (PDK1 or PDPK1) was overexpressed in HSCC. The overexpression was positively correlated lymph node metastasis, clinical stage, and distant metastasis and indicated poor outcome. Loss and gain-of-function revealed that PDK1 increased cell proliferation, migration and invasion in vitro as well as tumor growth and metastasis in vivo. Mechanically, PDK1 induced epithelial-mesenchymal transition and promoted metastasis by activating the Notch1 signaling pathway. We further illustrated that PDK1 bound with the Notch1 intracellular domain, thereby inhibiting its ubiquitin-mediated degradation in a protein kinase B (Akt-) independent manner. In summary, PDK1/Notch1 axis played an important role in HSCC metastasis, and this investigation provided a new perspective on potential therapeutic targets for HSCC.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Carcinoma de Células Escamosas/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas/genética , Receptor Notch1/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Anciano , Animales , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Hipofaríngeas/diagnóstico , Neoplasias Hipofaríngeas/mortalidad , Neoplasias Hipofaríngeas/patología , Metástasis Linfática , Masculino , Ratones Desnudos , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Receptor Notch1/metabolismo , Transducción de Señal , Análisis de Supervivencia , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cereb Cortex ; 30(1): 406-420, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31504280

RESUMEN

The six-layered neocortex consists of diverse neuron subtypes. Deeper-layer neurons originate from apical progenitors (APs), while upper-layer neurons are mainly produced by basal progenitors (BPs), which are derivatives of APs. As development proceeds, an AP generates two daughter cells that comprise an AP and a deeper-layer neuron or a BP. How the transition of APs to BPs is spatiotemporally regulated is a fundamental question. Here, we report that conditional deletion of phoshpoinositide-dependent protein kinase 1 (PDK1) in mouse developing cortex achieved by crossing Emx1Cre line with Pdk1fl/fl leads to a delayed transition of APs to BPs and subsequently causes an increased output of deeper-layer neurons. We demonstrate that PDK1 is involved in the modulation of the aPKC-Par3 complex and further regulates the asymmetric cell division (ACD). We also find Hes1, a downstream effecter of Notch signal pathway is obviously upregulated. Knockdown of Hes1 or treatment with Notch signal inhibitor DAPT recovers the ACD defect in the Pdk1 cKO. Thus, we have identified a novel function of PDK1 in controlling the transition of APs to BPs.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/fisiología , Corteza Cerebral/crecimiento & desarrollo , Células-Madre Neurales/fisiología , Neuronas/fisiología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
15.
J Cardiovasc Pharmacol ; 76(1): 77-85, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32398474

RESUMEN

Increasing evidence has confirmed that both long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) exert key roles in the pathogenesis of myocardial infarction (MI). Previous microarray assay results revealed that lncRNA LNC_000898 expression was significantly downregulated in acute MI. However, the specific function of LNC_000898 on MI is still unclear. Our study was aimed to explore the role of LNC_000898 on cardiac MI injury and investigate its underlying mechanism. The male C57BL/6 mouse was used as cardiac MI injury animal models, and neonatal mouse ventricular cardiomyocytes (NMCMs) exposed to hypoxia were used as an in vitro model. Quantitative real-time polymerase chain reaction analysis, Western blot analysis, Tunel immunofluorescence staining assay, and cardiac echocardiography measurement were conducted to detect corresponding indicators. The results indicated that LNC_000898 expression was downregulated in marginal tissue of MI and in NMCMs exposed to hypoxia. Overexpression of LNC_000898 decreased cardiomyocyte apoptosis both in vivo and in vitro. In addition, we elaborated that LNC_000898 exerts its inhibitory effect on apoptosis after MI through the miR-375/PDK1 axis. Through miR-375 overexpression or silencing PDK1, the biological effects of LNC_000898 on hypoxia-induced NMCM injury were partially reversed. These data not only demonstrate that LNC_000898 could protect the heart against MI injury by regulating miR-375/PDK1 but also provide a new understanding to better protection of MI injury through the LNC_000898/miR-375/PDK1 axis.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Apoptosis , MicroARNs/metabolismo , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , ARN Largo no Codificante/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Función Ventricular Izquierda , Remodelación Ventricular
16.
Diabetologia ; 62(7): 1225-1236, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31055616

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to elucidate the impact of 3'-phosphoinositide-dependent protein kinase-1 (PDPK1) in vascular endothelial cells on the maintenance of pancreatic beta cell mass and function. METHODS: Male vascular endothelial cell-specific Pdpk1-knockout mice (Tie2+/-/Pdpk1flox/flox mice) and their wild-type littermates (Tie2-/-/Pdpk1flox/flox mice; control) were used for this study. At 12 weeks of age, an IPGTT and OGTT were conducted. Pancreatic blood flow was measured under anaesthesia. Thereafter, islet blood flow was measured by the microsphere method. Mice were killed for islet isolation and further functional study and mRNA was extracted from islets. Pancreases were sampled for immunohistochemical analyses. RESULTS: During the IPGTT, the blood glucose level was comparable between knockout mice and control flox mice, although serum insulin level was significantly lower in knockout mice. During the OGTT, glucose tolerance deteriorated slightly in knockout mice, accompanied by a decreased serum insulin level. During an IPGTT after pre-treatment with exendin-4 (Ex-4), glucose tolerance was significantly impaired in knockout mice. In fact, glucose-stimulated insulin secretion of isolated islets from knockout mice was significantly reduced compared with control flox mice, and addition of Ex-4 revealed impaired sensitivity to incretin hormones in islets of knockout mice. In immunohistochemical analyses, both alpha and beta cell masses were significantly reduced in knockout mice. In addition, the CD31-positive area was significantly decreased in islets of knockout mice. The proportion of pimonidazole-positive islets was significantly increased in knockout mice. mRNA expression levels related to insulin biosynthesis (Ins1, Ins2, Mafa, Pdx1 and Neurod [also known as Neurod1]) and beta cell function (such as Gck and Slc2a2) were significantly decreased in islets of knockout mice. Microsphere experiments revealed remarkably reduced islet blood flow. In addition, mRNA expression levels of Hif1α (also known as Hif1a) and its downstream factors such as Adm, Eno1, Tpi1 (also known as Ets1), Hmox1 and Vegfa, were significantly increased in islets of knockout mice, indicating that islets of knockout mice were in a more hypoxic state than those of control flox mice. As a result, mRNA expression levels related to adaptive unfolded protein response and endoplasmic reticulum stress-related apoptotic genes were significantly elevated in islets of knockout mice. In addition, inflammatory cytokine levels were increased in islets of knockout mice. Electron microscopy revealed reduced endothelial fenestration and thickening of basal membrane of vascular endothelium in islets of knockout mice. CONCLUSIONS/INTERPRETATION: Vascular endothelial PDPK1 plays an important role in the maintenance of pancreatic beta cell mass and function by maintaining vascularity of pancreas and islets and protecting them from hypoxia, hypoxia-related endoplasmic reticulum stress, inflammation and distortion of capillary structure.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
17.
J Cell Mol Med ; 23(11): 7749-7761, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31475459

RESUMEN

Solamargine (SM) has been shown to have anti-cancer properties. However, the underlying mechanism involved remains undetermined. We showed that SM inhibited the growth of non-small cell lung cancer (NSCLC) cells, which was enhanced in cells with silencing of long non-coding RNA (lncRNA) HOX transcript antisense RNA (HOTAIR), while it overcame by overexpression of HOTAIR. In addition, SM increased the expression of miR-214-3p and inhibited 3-phosphoinositide-dependent protein kinase-1 (PDPK1) gene expression, which was strengthened by miR-214-3p mimics. Intriguingly, HOTAIR could directly bind to miR-214-3p and sequestered miR-214-3p from the target gene PDPK1. Intriguingly, overexpression of PDPK1 overcame the effects of SM on miR-214-3p expressions and neutralized the SM-inhibited cell growth. Similar results were observed in vivo. In summary, our results showed that SM-inhibited NSCLC cell growth through the reciprocal interaction between HOTAIR and miR-214-3p, which ultimately suppressed PDPK1 gene expression. HOTAIR effectively acted as a competing endogenous RNA (ceRNA) to stimulate the expression of target gene PDPK1. These complex interactions and feedback mechanisms contribute to the overall effect of SM. This unveils a novel molecular mechanism underlying the anti-cancer effect of SM in human lung cancer.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/antagonistas & inhibidores , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/genética , MicroARNs/genética , ARN Largo no Codificante/metabolismo , Alcaloides Solanáceos/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Humanos , Neoplasias Pulmonares/patología , Ratones Desnudos , MicroARNs/metabolismo , ARN Largo no Codificante/genética
18.
J Biol Chem ; 293(48): 18613-18623, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30305395

RESUMEN

Insulin promotes larval growth of insects by stimulating the synthesis of the steroid hormone 20-hydroxyecdysone (20E), which induces pupation and apoptosis. However, the mechanism underlying the coordinate regulation of insect pupation and apoptosis by these two functionally opposing hormones is still unclear. Here, using the lepidopteran insect and serious agricultural pest Helicoverpa armigera (cotton bollworm) as a model, we report that phosphoinositide-dependent kinase-1 (PDK1) and forkhead box O (FoxO) play key roles in these processes. We found that the transcript levels of the PDK1 gene are increased during the larval feeding stages. Moreover, PDK1 expression was increased by insulin, but repressed by 20E. dsRNA-mediated PDK1 knockdown in the H. armigera larvae delayed pupation and resulted in small pupae and also decreased Akt/protein kinase B expression and increased FoxO expression. Furthermore, the PDK1 knockdown blocked midgut remodeling and decreased 20E levels in the larvae. Of note, injecting larvae with 20E overcame the effect of the PDK1 knockdown and restored midgut remodeling. FoxO overexpression in an H. armigera epidermal cell line (HaEpi) did not induce apoptosis, but promoted autophagy and repressed cell proliferation. These results reveal cross-talk between insulin and 20E and that both hormones oppose each other's activities in the regulation of insect pupation and apoptosis by controlling PDK1 expression and, in turn, FoxO expression. We conclude that sufficiently high 20E levels are a key factor for inducing apoptosis during insect pupation.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Ecdisterona/fisiología , Proteínas de Insectos/metabolismo , Insulina/fisiología , Lepidópteros/crecimiento & desarrollo , Pupa/crecimiento & desarrollo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Apoptosis/genética , Autofagia/genética , Proliferación Celular/genética , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Larva/crecimiento & desarrollo , Fosforilación , ARN Mensajero/metabolismo
19.
J Cell Physiol ; 234(11): 20728-20741, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31004362

RESUMEN

Non-small-cell lung cancer (NSCLC) is an aggressive subtype of pulmonary carcinomas with high mortality. However, chemotherapy drug resistance and high recurrence rates hinder the curative effect of platinum-based first-line chemotherapy, which makes it urgent to develop new antitumor drugs for NSCLC. 9za, a new candidate drug synthesized by our research group, has been verified with potent antilung cancer activity in preliminary experiments. However, the underlying molecular mechanism of 9za remains largely vague. This work revealed that 9za could play important cytotoxic and proapoptotic roles in NSCLC cells. Moreover, 9za could induce autophagy and promote autophagy flux. Interestingly, the cytotoxic and proapoptotic roles were significantly dependent on 9za-induced cytoprotective autophagy. That is, the coadministration of 9za with an autophagy inhibitor such as chloroquine or 3-methyladenine exhibited increased cytotoxic and proapoptotic effects compared with 9za treatment alone. In addition, 9za exposure suppressed the phosphorylation of phosphoinositide-dependent protein kinase 1 (PDK1), protein kinase B (Akt), mammalian targets of rapamycin (mTOR), p70 S6 kinase, and 4E binding protein 1 by a dose-dependent way, manifesting that the Akt/mTOR axis was implicated in 9za-induced autophagy. In addition, the overexpression of PDK1 resulted in increased phosphorylation of PDK1 and Akt and blocking of 9za-mediated autophagy. These data showed that the PDK1/Akt/mTOR pathway was involved in 9za-induced autophagy. Hence, this work provides a theoretical basis for exploiting 9za as a new antilung cancer candidate drug and hints that the combination of 9za with an autophagy inhibitor is a feasible alternative approach for the therapy of NSCLC.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Benzodioxoles/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Indoles/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Compuestos de Anilina/química , Compuestos de Anilina/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Benzodioxoles/química , Benzodioxoles/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/química , Indoles/uso terapéutico , Estructura Molecular , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética
20.
J Cell Biochem ; 120(1): 126-134, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30218446

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Carcinoma de Células Renales/metabolismo , Glucólisis , Neoplasias Renales/metabolismo , MicroARNs/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/cirugía , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/patología , Neoplasias Renales/cirugía , MicroARNs/química , Imitación Molecular/genética , Consumo de Oxígeno , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA