Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 235(7-8): 5511-5524, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32017075

RESUMEN

There is now increasing evidence which suggests a key role for osteoblast apoptosis in the pathogenesis of postmenopausal osteoporosis. Here, we evaluated the role and mechanism of proteasome 26S subunit, ATPase (PSMC) 6, a protein that is highly expressed in bone. Gene expression pattern had been extracted based on database of Gene Expression Omnibus (GEO). GEO2R was employed for analyses, while the DAVID database was adopted to further analyze the gene ontology (GO) as well as Kyoto Encyclopedia of Genomes pathway (KEGG) enrichment. Then, the Search Tool Retrieval of Interacting Genes (STRING) was utilized to carry out interaction regulatory network for the top 200 differentially expressed genes (DEGs). A key gene, called PSMC6, was identified by Cytoscape 3.6.0. The OVX osteoporosis model was established in female C57BL/6 mice by full bilateral ovariectomy. According to our findings, PSMC6 gene knockout would elevate bone mineral density (BMD) and the phosphorylation level of PI3K protein and increased the protein level of cleaved caspase-3/-9 in OVX osteoporosis mice. Further, MTT, bromodeoxyuridine, and flow cytometry assays revealed that PSMC6 inhibition promoted the progression of cell cycle and cell proliferation, whereas, PSMC6 overexpression promoted the apoptosis and inhibited cell cycle progression and cell proliferation in vitro. Besides, we found that PI3K activation significantly decreased PSMC6-induced osteoblast apoptosis and promoted cell proliferation through regulating the protein levels of p53, cyclinD1, and cleaved caspase-3/9. In conclusion, PSMC6 aggravated the degree of OVX-induced osteoporosis by inhibiting the PI3K/AKT signal transduction pathway, thereby promoting the apoptosis of osteoblasts.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Apoptosis/genética , Osteoporosis Posmenopáusica/genética , Osteoporosis/genética , Complejo de la Endopetidasa Proteasomal/genética , ATPasas Asociadas con Actividades Celulares Diversas/antagonistas & inhibidores , Animales , Densidad Ósea/efectos de los fármacos , Proliferación Celular/genética , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/genética , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoporosis/etiología , Osteoporosis/fisiopatología , Osteoporosis Posmenopáusica/fisiopatología , Ovariectomía/efectos adversos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/genética
2.
Br J Cancer ; 122(9): 1298-1308, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32152504

RESUMEN

BACKGROUND: Akt signalling regulates glycolysis and drives the Warburg effect in cancer, thus decreased glucose utilisation is a pharmacodynamic marker of Akt inhibition. However, cancer cells can utilise alternative nutrients to glucose for energy such as lactate, which is often elevated in tumours together with increased acidity. We therefore hypothesised that lactic acidosis may confer resistance to Akt inhibition. METHODS: The effect of the pan-Akt inhibitor uprosertib (GSK2141795), on HCT116 and LS174T colon cancer cells was evaluated in the presence and absence of lactic acid in vitro. Expression of downstream Akt signalling proteins was determined using a phosphokinase array and immunoblotting. Metabolism was assessed using 1H nuclear magnetic resonance spectroscopy, stable isotope labelling and gas chromatography-mass spectrometry. RESULTS: Lactic acid-induced resistance to uprosertib was characterised by increased cell survival and reduced apoptosis. Uprosertib treatment reduced Akt signalling and glucose uptake irrespective of lactic acid supplementation. However, incorporation of lactate carbon and enhanced respiration was maintained in the presence of uprosertib and lactic acid. Inhibiting lactate transport or oxidative phosphorylation was sufficient to potentiate apoptosis in the presence of uprosertib. CONCLUSIONS: Lactic acidosis confers resistance to uprosertib, which can be reversed by inhibiting lactate transport or oxidative metabolism.


Asunto(s)
Acidosis Láctica/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Proteína Oncogénica v-akt/genética , Acidosis Láctica/genética , Acidosis Láctica/metabolismo , Acidosis Láctica/patología , Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Diaminas/farmacología , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Células HCT116 , Humanos , Ácido Láctico/farmacología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosforilación Oxidativa/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos
3.
Immunity ; 35(1): 34-44, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21683629

RESUMEN

Detection of microbial constituents by membrane associated and cytoplasmic pattern recognition receptors is the essence of innate immunity, leading to activation of protective host responses. However, it is still unclear how immune cells specifically respond to pathogenic bacteria. Using virulent and nonvirulent strains of Bacillus anthracis, we have shown that secretion of ATP by infected macrophages and the sequential activation of the P2X7 purinergic receptor and nucleotide binding oligomerization domain (NOD)-like receptors are critical for IL-1-dependent host protection from virulent B. anthracis. Importantly, lethal toxin produced by virulent B. anthracis blocked activation of protein kinases, p38 MAPK and AKT, resulting in opening of a connexin ATP release channel and induction of macrophage death. Prevention of cell death or ATP release through constitutive p38 or AKT activation interfered with inflammasome activation and IL-1ß production, thereby compromising antimicrobial immunity.


Asunto(s)
Carbunco/inmunología , Antígenos Bacterianos/metabolismo , Bacillus anthracis/inmunología , Toxinas Bacterianas/metabolismo , Inflamasomas/metabolismo , Macrófagos Peritoneales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Carbunco/microbiología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Bacillus anthracis/genética , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Células Cultivadas , Conexina 43/metabolismo , Inmunidad Innata/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/microbiología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Proteína Oncogénica v-akt/antagonistas & inhibidores , Receptores Purinérgicos P2X7/metabolismo , Virulencia/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
4.
Exp Mol Pathol ; 117: 104543, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32971114

RESUMEN

Long noncoding RNAs (lncRNAs) have critical roles in various malignancies. However, the specific expression and roles of lncRNA PTCSC1 in esophageal squamous cell carcinoma (ESCC) are still unknown. Here, we identified that lncRNA PTCSC1 was elevated in ESCC tissues and cell lines compared with adjacent noncancerous tissues and normal esophageal epithelial cell line, respectively. Enhanced expression of PTCSC1 facilitated ESCC cells proliferation and migration in vitro and ESCC xenograft growth in vivo. Conversely, deficiency of PTCSC1 suppressed ESCC cells proliferation and migration in vitro and ESCC tumor growth in vivo. Furthermore, PTCSC1 was found to activate Akt signaling in ESCC cells. Blocking Akt signaling with MK-2206 abolished the pro-proliferative and pro-migratory roles of PTCSC1. In summary, our findings demonstrated PTCSC1 as an oncogenic lncRNA in ESCC via activating Akt signaling and suggested that targeting PTCSC1 represents a promising therapeutic strategy against ESCC.


Asunto(s)
Proliferación Celular/genética , Carcinoma de Células Escamosas de Esófago/genética , Proteína Oncogénica v-akt/genética , ARN Largo no Codificante/genética , Animales , Apoptosis/genética , Movimiento Celular/genética , Progresión de la Enfermedad , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Xenoinjertos , Humanos , Masculino , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
5.
Molecules ; 25(20)2020 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080996

RESUMEN

A series of novel quinazolinone derivatives (2-13) was synthesized and examined for their cytotoxicity to HepG2, MCF-7, and Caco-2 in an MTT assay. Among these derivatives, compounds 4 and 9 exhibited significant cytotoxic activity against Caco-2, HepG2, and MCF-7 cancer cells. Compound 4 had more significant inhibitory effects than compound 9 on Caco-2, HepG2, and MCF-7 cell lines, with IC50 values of 23.31 ± 0.09, 53.29 ± 0.25, and 72.22 ± 0.14µM, respectively. The AKT pathway is one of human cancer's most often deregulated signals. AKT is also overexpressed in human cancers such as glioma, lung, breast, ovarian, gastric, and pancreas. A molecular docking study was performed to analyze the inhibitory action of newly synthetic quinazolinone derivatives against Homo sapiens AKT1 protein. Molecular docking simulations were found to be in accordance with in vitro studies, and hence supported the biological activity. The results suggested that compounds 4 and 9 could be used as drug candidates for cancer therapy via its potential inhibition of AKT1 as described by docking study.


Asunto(s)
Antineoplásicos/química , Neoplasias/tratamiento farmacológico , Proteína Oncogénica v-akt/genética , Quinazolinonas/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Células Hep G2 , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Estructura Molecular , Neoplasias/genética , Proteína Oncogénica v-akt/antagonistas & inhibidores , Quinazolinonas/síntesis química , Quinazolinonas/farmacología , Relación Estructura-Actividad
6.
J Virol ; 91(20)2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768870

RESUMEN

Progressive multifocal leukoencephalopathy (PML) is an often-fatal demyelinating disease of the central nervous system. PML results when oligodendrocytes within immunocompromised individuals are infected with the human JC virus (JCV). We have identified an oligodendrocyte precursor cell line, termed G144, that supports robust levels of JCV DNA replication, a central part of the JCV life cycle. In addition, we have determined that JC virus readily infects G144 cells. Furthermore, we have determined that JCV DNA replication in G144 cells is stimulated by myristoylated (i.e., constitutively active) Akt and reduced by the Akt-specific inhibitor MK2206. Thus, this oligodendrocyte-based model system will be useful for a number of purposes, such as studies of JCV infection, establishing key pathways needed for the regulation of JCV DNA replication, and identifying inhibitors of this process.IMPORTANCE The disease progressive multifocal leukoencephalopathy (PML) is caused by the infection of particular brain cells, termed oligodendrocytes, by the JC virus. Studies of PML, however, have been hampered by the lack of an immortalized human cell line derived from oligodendrocytes. Here, we report that the G144 oligodendrocyte cell line supports both infection by JC virus and robust levels of JCV DNA replication. Moreover, we have established that the Akt pathway regulates JCV DNA replication and that JCV DNA replication can be inhibited by MK2206, a compound that is specific for Akt. These and related findings suggest that we have established a powerful oligodendrocyte-based model system for studies of JCV-dependent PML.


Asunto(s)
Virus JC/fisiología , Oligodendroglía/virología , Proteína Oncogénica v-akt/metabolismo , Replicación Viral , Línea Celular , Replicación del ADN , ADN Viral , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/virología , Oligodendroglía/efectos de los fármacos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/química
7.
Toxicol Appl Pharmacol ; 356: 151-158, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30098992

RESUMEN

PIKfyve phosphoinositide kinase produces PtdIns(3,5)P2 and PtdIns5P and governs a myriad of cellular processes including cytoskeleton rearrangements and cell proliferation. The latter entails rigorous investigation since the cytotoxicity of PIKfyve inhibition is a potential therapeutic modality for cancer. Here we report the effects of two PIKfyve-specific inhibitors on the attachment/spreading and viability of mouse embryonic fibroblasts (MEFs) and C2C12 myoblasts. Importantly, 18-h treatment of adherent cells with YM201636 (800 nM) and apilimod (20 nM) in serum-containing culture media did not affect cell viability despite the presence of multiple cytoplasmic vacuoles, a hallmark of PIKfyve inhibition. Strikingly, at the same dose and duration the inhibitors caused excessive cytoplasmic vacuolation, initial suppression of cell attachment/spreading and subsequent marked detachment/death in serum-deprived cells. The remaining adherent cells under serum-deprived conditions had smaller surface area, lacked vinculin/actin-positive focal adhesions and displayed vacuoles occupying the entire cytoplasm. Serum or growth factors protected against PIKfyve inhibitor cytotoxicity. This protection required Akt activation evidenced by the abrogated beneficial effect of serum upon treatment with the clinically-relevant Akt inhibitor MK-2206. Moreover, Akt inhibition triggered cell detachment/death even in serum-fed adherent MEFs treated with apilimod. Intriguingly, BafilomycinA1 (H+-vacuolar ATPase inhibitor), which prevents the cytoplasmic vacuolation under PIKfyve perturbations, rescued all defects in attaching/spreading as well as in adherent cells under serum-starved or serum-fed conditions, respectively. Together, the results indicate that the cytotoxicity of PIKfyve inhibitors in MEFs and C2C12 myoblasts requires Akt suppression and excessive cytoplasmic vacuolation.


Asunto(s)
Antineoplásicos/farmacología , Citoplasma/efectos de los fármacos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Vacuolas/efectos de los fármacos , Aminopiridinas/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Recuento de Células , Muerte Celular/efectos de los fármacos , Citoplasma/ultraestructura , Inhibidores Enzimáticos/farmacología , Fibroblastos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Macrólidos/farmacología , Ratones , Mioblastos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas , Vacuolas/ultraestructura
8.
Med Sci Monit ; 24: 6093-6101, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30171812

RESUMEN

BACKGROUND Ovarian cancer is considered one of the lethal cancers responsible for high mortality and morbidity across the world. The prognosis and the survival rate of ovarian cancer is far from decent. Cysteine-rich 61 (Cyr61) also known as CCN1, is a member of CCN-family of growth factors, reported to be significantly overexpressed in several malignancies which include, but are not limited to, ovarian cancer. Recent studies have revealed that women with type 2 diabetes mellitus have an elevated risk of ovarian cancer. Hence, administration of an antidiabetic drug with anticancer effects such as metformin may act as an effective therapeutic regime against ovarian cancer. MATERIAL AND METHODS Cell viability and apoptosis were examined by MTT and Annexin V/PI double staining respectively. Cell migration was determined by Boyden Chamber assay. Transient knockdown of Cyr61 in ovarian cancer cells was achieved by transecting the cells with siRNA for Cyr61using Lipofectamine 2000. RESULTS Our results indicated that treatment of ovarian cancer cells with metformin caused significant downregulation of Cyr61 protein expression levels ultimately favoring apoptosis. Transient knockdown of Cyr61 resulted in the inhibition of cell proliferation and migration. This was also associated with the concomitant downregulation of pAkt and pmTOR confirming the role of Cyr61 as an upstream modulator of Akt signaling. Conversely the extracellular supplementation of recombinant Cyr61 attenuates the cytotoxic properties of metformin in ovarian cancer cells. CONCLUSIONS Taken together, we concluded that metformin exhibits anticancer effects and Cyr61 acts as a direct target for metformin in ovarian cancer cells.


Asunto(s)
Proteína 61 Rica en Cisteína/antagonistas & inhibidores , Metformina/farmacología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Proteína Oncogénica v-akt/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
9.
Biochem J ; 474(24): 4153-4170, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29046392

RESUMEN

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is triggered by BCR/ABL kinase. Recent efforts focused on the development of more potent tyrosine kinase inhibitors (TKIs) that also inhibit mutant tyrosine kinases such as nilotinib and dasatinib. Although major advances in the treatment of this aggressive disease with potent inhibitors of the BCR/ABL kinases, patients in remission frequently relapse due to drug resistance possibly mediated, at least in part, by compensatory activation of growth-signaling pathways and protective feedback signaling of leukemia cells in response to TKI treatment. Continuous activation of AKT/mTOR signaling and inactivation of p53 pathway were two mechanisms of TKI resistance. Here, we reported that nutlin-3 plus tanshinone IIA significantly potentiated the cytotoxic and apoptotic induction effects of imatinib by down-regulation of the AKT/mTOR pathway and reactivating the p53 pathway deeply in Ph+ ALL cell line. In primary samples from Ph+ ALL patients, nutlin-3 plus tanshinone IIA also exhibited synergetic cytotoxic effects with imatinib. Of note, three samples from Ph+ ALL patients harboring T315I mutation also showed sensitivity to the combined treatment of imatinib, nutlin-3 plus tanshinone IIA. In Ph+ ALL mouse models, imatinib combined with nutlin-3 plus tanshinone IIA also exhibited synergetic effects on reduction in leukemia burden. These results demonstrated that nutlin-3 plus tanshinone IIA combined TKI might be a promising treatment strategy for Ph+ ALL patients.


Asunto(s)
Abietanos/administración & dosificación , Mesilato de Imatinib/administración & dosificación , Imidazoles/administración & dosificación , Proteína Oncogénica v-akt/antagonistas & inhibidores , Piperazinas/administración & dosificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos Fitogénicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Genes p53/efectos de los fármacos , Genes p53/fisiología , Humanos , Células K562 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína Oncogénica v-akt/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento
10.
Proteomics ; 17(22)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28994205

RESUMEN

Mycobacterium tuberculosis (Mtb) cell wall glycolipid mannose-capped lipoarabinomannan (ManLAM) inhibits CD4+ T-cell activation by inhibiting proximal T-cell receptor (TCR) signaling when activated by anti-CD3. To understand the impact of ManLAM on CD4+ T-cell function when both the TCR-CD3 complex and major costimulator CD28 are engaged, we performed label-free quantitative MS and network analysis. Mixed-effect model analysis of peptide intensity identified 149 unique peptides representing 131 proteins that were differentially regulated by ManLAM in anti-CD3- and anti-CD28-activated CD4+ T cells. Crosstalker, a novel network analysis tool identified dysregulated translation, TCA cycle, and RNA metabolism network modules. PCNA, Akt, mTOR, and UBC were found to be bridge node proteins connecting these modules of dysregulated proteins. Altered PCNA expression and cell cycle analysis showed arrest at the G2M phase. Western blot confirmed that ManLAM inhibited Akt and mTOR phosphorylation, and decreased expression of deubiquitinating enzymes Usp9x and Otub1. Decreased NF-κB phosphorylation suggested interference with CD28 signaling through inhibition of the Usp9x-Akt-mTOR pathway. Thus, ManLAM induced global changes in the CD4+ T-cell proteome by affecting Akt-mTOR signaling, resulting in broad functional impairment of CD4+ T-cell activation beyond inhibition of proximal TCR-CD3 signaling.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Redes Reguladoras de Genes , Lipopolisacáridos/farmacología , Mycobacterium tuberculosis/metabolismo , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteómica/métodos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Ciclo Celular , Femenino , Manosa/química , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Proteína Oncogénica v-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
11.
Toxicol Appl Pharmacol ; 330: 65-73, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28711525

RESUMEN

Heat shock protein 90 (Hsp90) is a critically conserved molecular chaperone protein and promising therapeutic target for cancer treatment. In this study, platycodin D (PD), a saponin isolated from traditional Chinese herb Platycodonis Radix, was identified as a novel Hsp90 inhibitor. We verified that PD did not affect the ATPase activity of Hsp90. However, PD disrupted the co-chaperone interaction of Hsp90/cell division cycle protein 37 (Cdc37) and subsequently degraded multiple Hsp90 client proteins without the feedback increase of Hsp70. In different genotypes of non-small cell lung cancer cells, co-treatment with the mTOR inhibitor Everolimus and PD enhanced antiproliferation activity and apoptotic effect. The feedback survival signal upon mTOR inhibition was fully terminated by the co-administration with PD through reduced epidermal growth factor receptor (EGFR) and insulin growth factor 1 receptor (IGF1R) expression, suppressed AKT activity, and reinforced 4E-BP1 inhibition. Our results not only identified PD as a novel Hsp90 inhibitor by disrupting the protein-protein interaction of Hsp90/Cdc37 complex, but also provided mechanistic insights into the ineffectiveness of mTOR inhibitors and identified therapeutic strategy for cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/efectos de los fármacos , Chaperoninas/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Saponinas/toxicidad , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Triterpenos/toxicidad , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Everolimus/farmacología , Humanos , Inmunosupresores/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Proteína Oncogénica v-akt/antagonistas & inhibidores , Receptor IGF Tipo 1 , Receptores de Somatomedina/antagonistas & inhibidores
12.
Neurochem Res ; 42(5): 1354-1365, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28181071

RESUMEN

Regulation of various signalling (Ras-MAPK, PI3K and AKT) pathways by augmented activity of neurotrophic factors (NTFs) could prevent or halt the progress of dopaminergic loss in Parkinson's disease (PD). Various in vitro and in vivo experimental studies indicated anti-parkinsonic potential of asiatic acid (AA), a pentacyclic triterpene obtained from Centella asiatica. So the present study is designed to determine the neurotrophic effect of AA against 1-methyl 4-phenyl 1, 2, 3, 6-tetrahydropyridine hydrochloride/probenecid (MPTP/p) neurotoxicity in mice model of PD. AA treatment for 5 weeks significantly attenuated MPTP/p induced motor abnormalities, dopamine depletion and diminished expressions NTFs and tyrosine kinase receptors (TrKB). We further, revealed that AA treatment significantly inhibited the MPTP/p-induced phosphorylation of MAPK/P38 related proteins such as JNK and ERK. Moreover, AA treatment increased the phosphorylation of PI3K, Akt, GSK-3ß and mTOR, suggesting that AA activated PI3K/Akt/mTOR signalling pathway, which might be the cause of neuroprotection offered by AA. The present findings provided more elaborate in vivo evidences to support the neuroprotective effect of AA on dopaminergic neurons of chronic Parkinson's disease mouse model and the potential of AA to be developed as a possible new therapeutic target to treat PD.


Asunto(s)
Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/prevención & control , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Triterpenos Pentacíclicos/uso terapéutico , Probenecid/toxicidad , Serina-Treonina Quinasas TOR/metabolismo , Animales , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Intoxicación por MPTP/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/metabolismo , Triterpenos Pentacíclicos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Resultado del Tratamiento
13.
Neurochem Res ; 42(5): 1366-1374, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28247332

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) plays important roles in protecting the damaged or dying dopamine neurons in the animal models of Parkinson's disease (PD). This study was to determine the effect and mechanisms of GDNF on the apoptosis of neurons in 6-hydroxydopamine (6-OHDA) induced Parkinson's disease model of rats. Healthy male Sprague-Dawley rats (220-240 g) were randomly divided into six groups (n = 10). 6-OHDA was used to establish the PD rat model. Tyrosine hydroxylase (TH) immunohistochemistry was used to assess the neuron loss in 6-OHDA-lesioned rats. TUNEL and western blot were used to identify the effects and mechanisms of GDNF in the rat model of PD. The numbers of TH-positive neurons in the 6-OHDA-injected lesioned substantia nigra (SN) decreased significantly compared with the Sham group. GDNF treatment effectively ameliorated the apoptosis of neuronal cells in SN induced by 6-OHDA. In addition, GDNF significantly increased serine protein kinase B (Akt) and glycogen synthase kinase 3 beta (GSK3ß) phosphorylation induced by 6-OHDA. In contrast, application of LY294002 or triciribine reversed the roles of GDNF in PD models. The results implicated that the anti-apoptosis effects of GDNF in neurons might be mediated through PI3K/Akt/GSK3ß pathway. Therefore, GDNF may be a promising agent for PD treatment.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/administración & dosificación , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Neuronas/metabolismo , Proteína Oncogénica v-akt/metabolismo , Trastornos Parkinsonianos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Administración Intranasal , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Proteína Oncogénica v-akt/antagonistas & inhibidores , Oxidopamina/toxicidad , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/prevención & control , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología
14.
Exp Mol Pathol ; 103(3): 288-293, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29169802

RESUMEN

The PI3K/AKT/mTOR signaling pathway controls major cellular processes such as cell growth, proliferation and survival. Stimulation of this pathway leads to AKT phosphorylation and activation, resulting in phosphorylation of mTOR and myriad other targets. AKT upregulation has been implicated in thyroid cancer pathogenesis and is a candidate treatment target for patients with advanced disease that has not responded to traditional therapies. Here we evaluate a large series of benign and malignant thyroid tumors for AKT activity and intracellular distribution. We also deplete AKT from multiple thyroid cancer cell lines, including putative cancer stem cell lines, and measure the effect on proliferation and invasion in vitro. We show that active AKT has a predominantly nuclear distribution and its expression is highest in anaplastic thyroid carcinomas and papillary thyroid carcinomas, including encapsulated and invasive follicular variants. Depletion of AKT in thyroid carcinoma cell lines led to greatly reduced proliferative capacity and resulted in a reduction of invasive potential. A reduction in invasion was also observed in the cancer stem cell compartment. Targeting AKT activity in the clinical setting may slow the growth and spread of aggressive thyroid neoplasms, and target the tumor stem cell compartment.


Asunto(s)
Proliferación Celular/genética , Invasividad Neoplásica/genética , Proteína Oncogénica v-akt/genética , Neoplasias de la Tiroides/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica/patología , Células Madre Neoplásicas/patología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Neoplasias de la Tiroides/patología
15.
Acta Pharmacol Sin ; 38(7): 1048-1058, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28344324

RESUMEN

Diallyl trisulfide (DATS), a garlic organosulfide, has shown excellent chemopreventive potential. Cisplatin (DDP) is widely used to treat solid malignant tumors, but causing serious side effects. In the current study, we attempted to elucidate the chemopreventive mechanisms of DATS in human gastric cancer BGC-823 cells in vitro, and to investigate whether DATS could enhance the anti-tumor efficacy of DDP and improve quality of life in BGC-823 xenograft mice in vivo. Treatment with DATS (25-400 µmol/L) dose-dependently inhibited the viability of BGC-823 cells in vitro with an IC50 of 115.2±4.3 µmol/L after 24 h drug exposure. DATS (50-200 µmol/L) induced cell cycle arrest at G2/M phase in BGC-823 cells, which correlated with significant accumulation of cyclin A2 and B1. DATS also induced BGC-823 cell apoptosis, which was accompanied by the modulation of Bcl-2 family members and caspase cascade activation. In BGC-823 xenograft mice, administration of DATS (20-40 mg·kg-1·d-1, ip) dose-dependently inhibited tumor growth and markedly reduced the number of Ki-67 positive cells in tumors. Interestingly, combined administration of DATS (30 mg·kg-1·d-1, ip) with DDP (5 mg/kg, every 5 d, ip) exhibited enhanced anti-tumor activity with fewer side effects. We showed that treatment of BGC-823 cells with DATS in vitro and in vivo significantly activated kinases such as p38 and JNK/MAPK and attenuated the Nrf2/Akt pathway. This study provides evidence that DATS exerts anticancer effects and enhances the antitumor efficacy of DDP, making it a novel candidate for adjuvant therapy for gastric cancer.


Asunto(s)
Compuestos Alílicos/farmacología , Antineoplásicos/farmacología , Cisplatino/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Sulfuros/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/metabolismo , Neoplasias Gástricas/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Neurochem ; 137(5): 701-13, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26926143

RESUMEN

Recent studies have demonstrated brain insulin signaling impairment and mitochondrial dysfunction in diabetes. Hyperinsulinemia and hyperlipidemia arising from diabetes have been linked to neuronal insulin resistance, and hyperglycemia induces peripheral sensory neuronal impairment and mitochondrial dysfunction. However, how brain glucose at diabetic conditions elicits cortical neuronal insulin signaling impairment and mitochondrial dysfunction remains unknown. In the present study, we cultured primary cortical neurons with high glucose levels and investigated the neuronal mitochondrial function and insulin response. We found that mitochondrial function was declined in presence of 10 mmol/L glucose, prior to the depression of AKT signaling in primary cortical neurons. We further demonstrated that the cerebral cortex of db/db mice exhibited both insulin resistance and loss of mitochondrial complex components. Moreover, we found that adenosine monophosphate-activated protein kinase (AMPK) inactivation is involved in high glucose-induced mitochondrial dysfunction and insulin resistance in primary cortical neurons and neuroblastoma cells, as well as in cerebral cortex of db/db mice, and all these impairments can be rescued by mitochondrial activator, resveratrol. Taken together, our results extend the finding that high glucose (≥10 mmol/L) comparable to diabetic brain extracellular glucose level leads to neuronal mitochondrial dysfunction and resultant insulin resistance, and targeting mitochondria-AMPK signaling might be a promising strategy to protect against diabetes-related neuronal impairment in central nerves system. We found that high glucose (≥10 mmol/L), comparable to diabetic brain extracellular glucose level, leads to neuronal mitochondrial dysfunction and resultant insulin resistance in an AMPK-dependent manner, and targeting mitochondria-AMPK signaling might be a promising strategy to protect against diabetes-related neuronal impairment in central nerves system.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucosa/toxicidad , Resistencia a la Insulina/fisiología , Mitocondrias/metabolismo , Neuronas/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
17.
J Pharmacol Exp Ther ; 357(2): 345-56, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26941172

RESUMEN

Interaction of cardiac steroids (CS) with the Na(+), K(+)-ATPase elicits, in addition to inhibition of the enzyme's activity, the activation of intracellular signaling such as extracellular signal-regulated (ERK) and protein kinase B (Akt). We hypothesized that the activities of these pathways are involved in CS-induced increase in heart contractility. This hypothesis was tested using in vivo and ex vivo wild type (WT) and sarcoplasmic reticulum Ca(2+) atpase1a-deficient zebrafish (accordion, acc mutant) experimental model. Heart contractility was measured in vivo and in primary cardiomyocytes in WT zebrafish larvae and acc mutant. Ca(2+) transients were determined ex vivo in adult zebrafish hearts. CS dose dependently augmented the force of contraction of larvae heart muscle and cardiomyocytes and increased Ca(2+) transients in WT but not in acc mutant. CS in vivo increased the phosphorylation rate of ERK and Akt in the adult zebrafish heart of the two strains. Pretreatment of WT zebrafish larvae or cardiomyocytes with specific MAPK inhibitors completely abolished the CS-induced increase in contractility. On the contrary, pretreatment with Akt inhibitor significantly enhanced the CS-induced increase in heart contractility both in vivo and ex vivo without affecting CS-induced Ca(2+) transients. Furthermore, pretreatment of the acc mutant larvae or cardiomyocytes with Akt inhibitor restored the CS-induced increase in heart contractility also without affecting Ca(2+) transients. These results support the notion that the activity of MAPK pathway is obligatory for CS-induced increases in heart muscle contractility. Akt activity, on the other hand, plays a negative role, via Ca(2+) independent mechanisms, in CS action. These findings point to novel potential pharmacological intervention to increase CS efficacy.


Asunto(s)
Cardiotónicos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Proteína Oncogénica v-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esteroides/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Larva , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mutación , Miocitos Cardíacos/efectos de los fármacos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/deficiencia , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
18.
Indian J Med Res ; 143(2): 145-59, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27121513

RESUMEN

Cellular resistance in tumour cells to different therapeutic approaches has been a limiting factor in the curative treatment of cancer. Resistance to therapeutic radiation is a common phenomenon which significantly reduces treatment options and impacts survival. One of the mechanisms of acquiring resistance to ionizing radiation is the overexpression or activation of various oncogenes like the EGFR (epidermal growth factor receptor), RAS (rat sarcoma) oncogene or loss of PTEN (phosphatase and tensin homologue) which in turn activates the phosphatidyl inositol 3-kinase/protein kinase B (PI3-K)/AKT pathway responsible for radiation resistance in various tumours. Blocking the pathway enhances the radiation response both in vitro and in vivo. Due to the differential activation of this pathway (constitutively activated in tumour cells and not in the normal host cells), it is an excellent candidate target for molecular targeted therapy to enhance radiation sensitivity. In this regard, HIV protease inhibitors (HPIs) known to interfere with PI3-K/AKT signaling in tumour cells, have been shown to sensitize various tumour cells to radiation both in vitro and in vivo. As a result, HPIs are now being investigated as possible radiosensitizers along with various chemotherapeutic drugs. This review describes the mechanisms by which PI3-K/AKT pathway causes radioresistance and the role of HIV protease inhibitors especially nelfinavir as a potential candidate drug to target the AKT pathway for overcoming radioresistance and its use in various clinical trials for different malignancies.


Asunto(s)
Inhibidores de la Proteasa del VIH/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Proteína Oncogénica v-akt/genética , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Receptores ErbB/biosíntesis , Humanos , Neoplasias/patología , Proteína Oncogénica p21(ras)/biosíntesis , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosfohidrolasa PTEN/biosíntesis , Fosfatidilinositol 3-Quinasas/genética , Radiación Ionizante , Transducción de Señal/efectos de los fármacos
19.
BMC Cancer ; 15: 807, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26503030

RESUMEN

BACKGROUND: Malignant melanoma is an aggressive type of skin cancer, and despite recent advances in treatment, the survival rate of the metastatic form remains low. Nifuroxazide analogues are drugs based on the substitution of the nitrofuran group by benzofuroxan, in view of the pharmacophore similarity of the nitro group, improving bioavailability, with higher intrinsic activity and less toxicity. Benzofuroxan activity involves the intracellular production of free-radical species. In the present work, we evaluated the antitumor effects of different benzofuroxan derivatives in a murine melanoma model. METHODS: B16F10-Nex2 melanoma cells were used to investigate the antitumor effects of Benzofuroxan derivatives in vitro and in a syngeneic melanoma model in C57Bl/6 mice. Cytotoxicity, morphological changes and reactive oxygen species (ROS) were assessed by a diphenyltetrasolium reagent, optical and fluorescence microscopy, respectively. Annexin-V binding and mitochondrial integrity were analyzed by flow cytometry. Western blotting and colorimetry identified cell signaling proteins. RESULTS: Benzofuroxan N-Br and N-I derivatives were active against murine and human tumor cell lines, exerting significant protection against metastatic melanoma in a syngeneic model. N-Br and N-I induce apoptosis in melanoma cells, evidenced by specific morphological changes, DNA condensation and degradation, and phosphatidylserine translocation in the plasma membrane. The intrinsic mitochondrial pathway in B16F10-Nex2 cells is suggested owing to reduced outer membrane potential in mitochondria, followed by caspase -9, -3 activation and cleavage of PARP. The cytotoxicity of N-Br and N-I in B16F10-Nex2 cells is mediated by the generation of ROS, inhibited by pre-incubation of the cells with N-acetylcysteine (NAC). The induction of ROS by N-Br and N-I resulted in the inhibition of AKT activation, an important molecule related to tumor cell survival, followed by upregulation of BIM. CONCLUSION: We conclude that N-Br and N-I are promising agents aiming at cancer treatment. They may be useful in melanoma therapy as inducers of intrinsic apoptosis and by exerting significant antitumor activity against metastatic melanoma, as presently shown in syngeneic mice.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Benzoxazoles/química , Benzoxazoles/uso terapéutico , Melanoma Experimental/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteína 11 Similar a Bcl2 , Benzoxazoles/farmacología , Línea Celular Tumoral , Células HeLa , Humanos , Masculino , Melanoma Experimental/tratamiento farmacológico , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
20.
Mol Cell Biochem ; 400(1-2): 287-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25476740

RESUMEN

CXCL13, an inflammatory factor in the microenvironment, plays a vital role in the progression of inflammatory diseases and tumors. CXCL13 and its receptor CXCR5 have been reported to be associated with poor prognosis of advanced colon cancer. However, the molecular mechanisms of CXCL13-CXCR5 axis in colon cancer remain elusive. The aim of this study was to investigate the role of CXCR5-CXCL13 axis in the growth and invasion of colon cancer cells. Our results showed that CXCL13 promoted the growth, migration, and matrigel invasion of colon cancer cells. Furthermore, CXCL13 increased the expression and secretion of MMP-13, and stimulated the activation of PI3K/AKT pathway. After knockdown of CXCR5 by siRNA, the biological functions of colon cancer cells regulated by CXCL13 were significantly inhibited. In addition, inhibition of PI3K/AKT pathway by specific inhibitor LY294002 suppressed the CXCL13-mediated growth, migration, and invasion of colon cancer cells. Together, our findings suggest that CXCL13-CXCR5 axis promotes the growth, migration, and invasion of colon cancer cells, probably via PI3K/AKT pathway. Thus, CXCL13 may be a useful biomarker for the detection and treatment of colon cancer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Quimiocina CXCL13/biosíntesis , Neoplasias del Colon/genética , Receptores CXCR5/biosíntesis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular/genética , Quimiocina CXCL13/genética , Cromonas , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Morfolinas , Invasividad Neoplásica/genética , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Interferente Pequeño , Receptores CXCR5/genética , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA