Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 138
1.
Int J Biol Sci ; 17(7): 1808-1820, 2021.
Article En | MEDLINE | ID: mdl-33994864

Increasing evidence reveals that the Rho GTPase-activating protein is a crucial negative regulator of Rho family GTPase involved in tumorigenesis. The Rho GTPase-activating protein 25 (ARHGAP25) has been shown to specifically inactivate the Rho family GTPase Rac1, which plays an important role in pancreatic adenocarcinoma (PAAD) progression. Therefore, here we aimed to clarify the expression and functional role of ARHGAP25 in PAAD. The ARHGAP25 expression was lower in PAAD tissues than that in normal pancreatic tissues based on bioinformatics analysis and immunohistochemistry staining. Overexpression of ARHGAP25 inhibited cell growth of AsPC-1 human pancreatic cancer cells in vitro, while opposite results were observed in BxPC-3 human pancreatic cancer cells with ARHGAP25 knockdown. Consistently, in vivo tumorigenicity assays also confirmed that ARHGAP25 overexpression suppressed tumor growth. Mechanically, overexpression of ARHGAP25 inactivated AKT/mTOR signaling pathway by regulating Rac1/PAK1 signaling, which was in line with the results from the Gene set enrichment analysis on The Cancer Genome Atlas dataset. Furthermore, we found that ARHGAP25 reduced HIF-1α-mediated glycolysis in PAAD cells. Treatment with PF-04691502, a dual PI3K/mTOR inhibitor, hampered the increased cell growth and glycolysis due to ARHGAP25 knockdown in PAAD cells. Altogether, these results conclude that ARHGAP25 acts as a tumor suppressor by inhibiting the AKT/mTOR signaling pathway, which might provide a therapeutic target for PAAD.


Adenocarcinoma/genetics , Carcinogenesis/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Oncogene Protein v-akt/genetics , Pancreatic Neoplasms/genetics , TOR Serine-Threonine Kinases/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Female , GTPase-Activating Proteins/biosynthesis , Glycolysis/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasms, Experimental , Oncogene Protein v-akt/biosynthesis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/biosynthesis
2.
FASEB J ; 34(2): 2075-2086, 2020 02.
Article En | MEDLINE | ID: mdl-31907982

In the free-living nematode Caenorhabditis elegans, the serine/threonine-specific protein kinase, AKT, is known to play a key role in dauer formation, life-span, and stress-resistance through the insulin-like signaling pathway. Although the structure and function of AKT-coding genes of C. elegans are understood, this is not the case for homologous genes in parasitic nematodes. In the present study, we explored a C. elegans akt-1 gene homolog in the parasitic nematode Haemonchus contortus, investigated its transcript isoforms (Hc-akt-1a and Hc-akt-1b), and studied expression and function using both homologous and heterologous functional genomic tools. In C. elegans, we showed that the predicted promoter of Hc-akt-1 drives substantial expression in ASJ neurons of the N2 (wild-type) strain. In H. contortus (Haecon-5 stain), RNAi (soaking) led to a significantly decreased transcript abundance for both Hc-akt-1a and Hc-akt-1b, and reduced larval development in larval stages in vitro. Chemical inhibition was also shown to block larval development. Taken together, the evidence from this study points to a key functional role for Hc-akt-1 in H. contortus.


Gene Expression Regulation, Enzymologic , Haemonchus/growth & development , Helminth Proteins/biosynthesis , Neurons/enzymology , Oncogene Protein v-akt/biosynthesis , Animals , Haemonchus/genetics , Helminth Proteins/genetics , Isoenzymes/biosynthesis , Isoenzymes/genetics , Larva/genetics , Larva/growth & development , Oncogene Protein v-akt/genetics
3.
Eur Rev Med Pharmacol Sci ; 23(10): 4285-4292, 2019 May.
Article En | MEDLINE | ID: mdl-31173300

OBJECTIVE: PTEN-PI3K/AKT signaling pathway is widely involved in the regulation of cell proliferation, cell cycle, apoptosis, and invasion. Resveratrol (Resv) is a natural botanical ingredient involved in several biological activities. It is still unclear in terms of whether Resv may exert anti-leukemia effects by regulating the PTEN-PI3K/AKT pathway. This study investigated the effect of Resv on leukemia cell proliferation and apoptosis by regulating PTEN-PI3K/AKT pathway. PATIENTS AND METHODS: Human normal peripheral blood PBMC cells, and human acute promyelocytic leukemia (APL) cell line NB-4 and HL-60 cells were cultured in vitro. Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to detect Phosphatase and tensin homolog (PTEN) mRNA expression. Western blot was adopted to test PTEN protein expression. HL-60 and NB-4 cells were treated with 0, 5, 10, and 20 µM Resv, respectively. Cell proliferation was analyzed by cell counting kit8 (CCK-8) assay. The level of caspase-3 was measured by Western blot. HL-60 cells were divided into control group, 20 µM Resv treatment group, and Resv+PTEN inhibitor SF1670 group. Cell apoptosis was determined by flow cytometry. Cell proliferation was assessed by EdU staining. RESULTS: Compared with peripheral blood mononuclear cell (PBMC), PTEN mRNA and protein levels were significantly decreased in NB-4 and HL-60 cells. Resv significantly inhibited the proliferation activity in HL-60 and NB-4 cells, and increased the activity of caspase-3. Resv treatment up-regulated the expression of PTEN and reduced the expression of p-AKT protein in HL-60 cells. However, Resv treatment markedly suppressed the proliferation of HL-60 and induced apoptosis. SF1670 treatment in the presence of Resv significantly antagonized the down-regulation of p-AKT protein expression induced by Resv, resulting in decreased apoptosis and enhanced cell proliferation. CONCLUSIONS: Resv can up-regulate PTEN expression and inhibit the activity of PI3K/AKT pathway to play an anti-leukemia effect through suppressing cell proliferation and inducing apoptosis.


Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Leukemia/drug therapy , Oncogene Protein v-akt/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Resveratrol/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line, Tumor , HL-60 Cells , Humans , Leukemia/pathology , Oncogene Protein v-akt/biosynthesis , PTEN Phosphohydrolase/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis
4.
Bosn J Basic Med Sci ; 19(1): 52-59, 2019 Feb 12.
Article En | MEDLINE | ID: mdl-29924958

Triple-negative breast cancer (TNBC) is the leading cause of cancer-related death in women. Previous studies indicated that miR-361-5p was downregulated in breast cancer, however, the exact effect of miR-361-5p on TNBC requires further investigation. In the present study, we investigated whether miR-361-5p can act as a tumor suppressor by targeting required for cell differentiation 1 homolog (RQCD1) and inhibiting epidermal growth factor receptor (EGFR)/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway in TNBC. The expression of miR-361-5p and RQCD1 was determined by quantitative reverse transcription PCR (qRT-PCR) and/or western blot in TNBC and the adjacent tissues. miR-361-5p mimics were constructed and transfected to TNBC cell line MDA-MB-231. Cells were divided into three groups: blank control group, miRNA mimic negative control (NC) group, and miR-361-5p mimics group. Expression of miR-361-5p, mRNA and protein expression of PI3K, Akt, EGFR, phosphorylated (p)-EGFR/PI3K/Akt, and protein expression of RQCD1 and matrix metallopeptidase 9 (MMP-9) in MDA-MB-231 were measured by qRT-PCR/western blot after transfection. Cell viability was determined by CCK-8 assay. Cell migration and invasion ability were evaluated by scratch and transwell assay, respectively. miR-361-5p target gene was determined by bioinformatics analysis and luciferase reporter assay. RQCD1 was identified as a target of miR-361-5p by TargetScan and confirmed by luciferase reporter assay. Downregulated miR-361-5p and upregulated RQCD1 were observed in TNBC tissues. Expression of EGFR, PI3K, Akt and MMP-9 was inhibited in cells treated with miR-361-5p mimics. Transfection of miR-361-5p mimics also inhibited the phosphorylation of EGFR, PI3K, and Akt. Suppressed cell viability, migration, and invasion was found in miR-361-5p mimics groups. Our results indicated that overexpression of miR-361-5p might act as a suppressor in TNBC by targeting RQCD1 to inhibit the EGFR/PI3K/Akt signaling pathway.


Breast Neoplasms/metabolism , Breast Neoplasms/pathology , MicroRNAs/biosynthesis , Signal Transduction/genetics , Transcription Factors/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Adult , Cell Line, Tumor , Cell Movement/genetics , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/genetics , Transcription Factors/genetics
5.
Toxicol Appl Pharmacol ; 356: 224-234, 2018 10 01.
Article En | MEDLINE | ID: mdl-30138657

Patulin (PAT), a mycotoxin, is a natural contaminant that is produced by certain species of Penicillium, Aspergillus and Byssochlamys. The major contamination of PAT is in apple and apple based products. PAT is known to cause glutathione depletion, oxidative DNA damage and cell proliferation. Recently, in vitro studies have indicated that PAT can also increase the intestinal epithelial permeability, modulate tight junctions and decrease transepithelial electrical resistance. Nonetheless, no previous study has evaluated the mechanisms responsible for PAT-induced intestinal toxicity or its relevance to the in vivo situation. Here, Wistar rats were orally treated with 100 µg/kg body weight (b.wt.) of PAT, either alone or along with 100 mg/kg b. wt. of celecoxib for 3 days. We found that PAT exposure led to significantly higher levels of PGE2 in serum and intestinal tissue and high expression of COX-2 and Ki-67 compared to controls. Interestingly, our results showed that celecoxib treatment could decrease the PAT-induced PGE2 and reduce the PAT-induced intestinal damage. To study the mechanistic aspect, normal rat intestinal epithelial cells (IEC-6) were treated with non-toxic concentrations (100 nM, 250 nM and 500 nM) of PAT for 6 h. It was observed that PAT exposure caused enhanced proliferation, higher expression of COX-2, and EP2 and EP4 receptors, along with increased PGE2 secretion. Additionally, PAT exposure caused enhanced Akt expression, which in turn inhibits GSK-3ß and stabilizes ß-catenin. Overall, our study suggests that the COX-2/EP2-EP4/ß-catenin signaling cascades are involved in the regulation of PAT-induced intestinal cell proliferation and inflammation.


Cell Proliferation/drug effects , Cyclooxygenase 2/drug effects , Enteritis/drug therapy , Intestines/cytology , Intestines/drug effects , Patulin/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/drug effects , Signal Transduction/genetics , beta Catenin/drug effects , Animals , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/blood , Enteritis/physiopathology , Epithelial Cells/drug effects , Glycoproteins , Male , Oncogene Protein v-akt/biosynthesis , Plant Proteins , Rats , Rats, Wistar , Receptors, Prostaglandin E, EP4 Subtype/drug effects , Signal Transduction/physiology
6.
Biol Pharm Bull ; 41(3): 394-398, 2018.
Article En | MEDLINE | ID: mdl-29491216

Androgens are key regulators that play a critical role in the male reproductive system and have anabolic effects on bone mineral density and skeletal muscle mass. We have previously reported that YK11 is a novel selective androgen receptor modulator (SARM) and induces myogenic differentiation and selective gene regulation. In this study, we show that treatment of YK11 and dihydrotestosterone (DHT) accelerated cell proliferation and mineralization in MC3T3-E1 mouse osteoblast cells. Further, YK11-treated cells increased osteoblast specific differentiation markers, such as osteoprotegerin and osteocalcin, compared to untreated cells. These observations were attenuated by androgen receptor (AR) antagonist treatment. To clarify the effect of YK11, we investigated rapid non-genomic signaling by AR. The phosphorylated Akt protein level was increased by YK11 and DHT treatment, suggesting that YK11 activates Akt-signaling via non-genomic signaling of AR. Because it is known Akt-signaling is a key regulator of androgen-mediated osteoblast differentiation, YK11 has osteogenic activity as well as androgen.


Androgens/pharmacology , Norpregnadienes/pharmacology , Osteoblasts/drug effects , 3T3 Cells , Animals , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mice , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Osteogenesis/drug effects , Up-Regulation/drug effects
7.
Life Sci ; 196: 1-8, 2018 Mar 01.
Article En | MEDLINE | ID: mdl-28751159

AIMS: Acute lung injury (ALI) is associated with excessive mortality and lacks appropriate therapy. Ghrelin is a novel peptide that protects the lung against ALI. This study aimed to investigate whether endoplasmic reticulum stress (ERS) mediates the protective effect of ghrelin on ALI. MAIN METHODS: We used a rat oleic acid (OA)-induced ALI model. Pulmonary impairment was detected by hematoxylin and eosin (HE) staining, lung mechanics, wet/dry weight ratio, and arterial blood gas analysis. Plasma and lung content of ghrelin was examined by ELISA, and mRNA expression was measured by quantitative real-time PCR. Protein levels were detected by western blot. KEY FINDINGS: Rats with OA treatment showed significant pulmonary injury, edema, inflammatory cellular infiltration, cytokine release, hypoxia and CO2 retention as compared with controls. Plasma and pulmonary content of ghrelin was reduced in rats with ALI, and mRNA expression was downregulated. Ghrelin (10nmol/kg) treatment ameliorated the above symptoms, but treatment with the ghrelin antagonists D-Lys3 GHRP-6 (1µmol/kg) and JMV 2959 (6mg/kg) exacerbated the symptoms. ERS induced by OA was prevented by ghrelin and augmented by ghrelin antagonist treatment. The ERS inducer, tunicamycin (Tm) prevented the ameliorative effect of ghrelin on ALI. The decreased ratio of p-Akt and Akt induced by OA was improved by ghrelin treatment, and was further exacerbated by ghrelin antagonists. SIGNIFICANCE: Ghrelin protects against ALI by inhibiting ERS. These results provide a new target for prevention and therapy of ALI.


Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Endoplasmic Reticulum Stress/drug effects , Ghrelin/therapeutic use , Oleic Acid , Acute Lung Injury/physiopathology , Animals , Blood Gas Analysis , Bronchoalveolar Lavage Fluid , Carbon Dioxide/metabolism , Ghrelin/antagonists & inhibitors , Male , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Organ Size , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Tunicamycin/pharmacology
8.
Life Sci ; 192: 1-8, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-29138115

AIMS: Perinatal asphyxia (PA) often results in hypoxic-ischemic encephalopathy (HIE) in term neonates. Introduction of therapeutic hypothermia improved HIE outcome, but further neuroprotective therapies are still warranted. The present study sought to determine the feasibility of the activation of the cytoprotective PI-3-K/Akt and the MAPK/ERK signaling pathways in the subacute phase of HIE development in a translational newborn pig PA/HIE model. MAIN METHODS: Phosphorylated and total levels of Akt and ERK were determined by Western blotting in brain samples obtained from untreated naive, time control, and PA/HIE animals at 24-48h survival (n=3-3-6,respectively). PA (20min) was induced in anesthetized piglets by ventilation with a hypoxic/hypercapnic (6%O220%CO2) gas mixture. Furthermore, we studied the effect of topically administered specific Akt1/2 and MAPK/ERK kinase inhibitors on Akt and ERK phosphorylation (n=4-4) in the cerebral cortex under normoxic conditions. KEY FINDINGS: PA resulted in significant neuronal injury shown by neuropathology assessment of haematoxylin/eosin stained sections. However, there were no significant differences among the groups in the high phosphorylation levels of both ERK and Akt in the cerebral cortex, hippocampus and subcortical structures. However, the Akt1/2 and MAPK/ERK kinase inhibitors significantly reduced cerebrocortical Akt and ERK phosphorylation within 30min. SIGNIFICANCE: The major finding of the present study is that the PI-3-K/Akt and the MAPK/ERK signaling pathways appear to be constitutively active in the piglet brain, and this activation remains unaltered during HIE development. Thus, neuroprotective strategies aiming to activate these pathways to limit apoptotic neuronal death may offer limited efficacy in this translational model.


Cerebral Cortex/metabolism , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Oncogene Protein v-akt/biosynthesis , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Blood Pressure/drug effects , Cerebral Cortex/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Heart Rate/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , MAP Kinase Signaling System/drug effects , Male , Neurons/pathology , Oncogene Protein v-akt/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Swine
9.
J Pharmacol Exp Ther ; 363(3): 428-443, 2017 12.
Article En | MEDLINE | ID: mdl-29021381

Despite encouraging clinical results with sorafenib monotherapy in patients with KRAS-mutant non-small-cell lung cancer (NSCLC), the overall survival benefit of this drug is limited by the inevitable development of acquired resistance. The exact mechanism underlying acquired sorafenib resistance in KRAS-mutant NSCLC is unclear. In this study, the mechanism of acquired sorafenib resistance was explored using a biologically relevant xenograft model, which was established by using the A549 human lung adenocarcinoma cell line and an in vivo-derived, sorafenib-resistant A549 subline (A549/SRFres). Results from the initial study demonstrated that sorafenib treatment significantly decreased E-cadherin (P < 0.05) levels but significantly increased matrix metallopeptidase 9 (MMP9) levels (P < 0.01) in A549/SRFres tumors, whereas expression levels of phospho-protein kinase B (AKT), phospho-focal adhesion kinase (FAK), and phospho-Src were elevated in sorafenib-treated A549 and A549/SRFres tumors. We next examined whether concomitant dasatinib treatment could overcome acquired sorafenib resistance by blocking the FAK/Src escape route that mediates resistance. Despite the observed in vitro synergy between sorafenib and dasatinib, the in vivo antitumor effect of half-dose sorafenib-dasatinib combination therapy was inferior to that of the full-dose sorafenib treatment. Although the sorafenib-dasatinib combination effectively inhibited Src and AKT phosphorylation, it did not block the Y576/577-FAK phosphorylation, nor did it decrease vimentin protein expression; unexpectedly, it increased Y397-FAK phosphorylation and MMP9 protein expression in tumors. These results suggest that acquired sorafenib resistance in KRAS-mutant A549 xenografts involves the compensatory activation of FAK and Src, and Src inhibition alone is insufficient to diminish sorafenib-promoted epithelial-mesenchymal transition process and invasive potentials in tumors.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Focal Adhesion Protein-Tyrosine Kinases/genetics , Genes, src/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , A549 Cells , Animals , Dasatinib/pharmacology , Drug Synergism , Enzyme Activation , Humans , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Nude , Niacinamide/pharmacology , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Sorafenib , Xenograft Model Antitumor Assays
10.
Tumour Biol ; 39(7): 1010428317701441, 2017 Jul.
Article En | MEDLINE | ID: mdl-28705116

Alex3, is a newly identified mitochondrial protein, regulates mitochondrial dynamics and is involved in neural development. However, its expression pattern and clinicopathological relevance in human tumors are still unclear. In this study, Immunohistochemistry assay was performed in 109 cases of lung cancer samples and found that Alex 3 expression in lung cancer tissues was significantly lower than adjacent normal lung tissues (28.4% vs 52.6%, p < 0.001). Sequent statistical analysis indicated that negative Alex3 expression was significantly associated with advanced tumor-node-metastasis stages (p = 0.001), positive lymph node metastasis (p = 0.005), and poor prognosis (p = 0.008). After overexpression of Alex3, levels of p-AKT and Slug were downregulated, while level of E-cadherin was upregulated, which results in the inhibition of invasion and migration ability of lung cancer cells. In conclusion, reduction of Alex3 correlates with the development of non-small cell lung cancer and predicts adverse clinical outcome of non-small cell lung cancer patients. The effect of Alex3 on inhibiting invasion and migration may attribute to upregulation of E-cadherin expression through AKT-Slug pathway inactivation.


Armadillo Domain Proteins/genetics , Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Mitochondrial Proteins/genetics , Oncogene Protein v-akt/genetics , Snail Family Transcription Factors/genetics , Adult , Aged , Armadillo Domain Proteins/biosynthesis , Cadherins/biosynthesis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Middle Aged , Mitochondrial Proteins/biosynthesis , Neoplasm Invasiveness/genetics , Oncogene Protein v-akt/biosynthesis , Signal Transduction , Snail Family Transcription Factors/biosynthesis
11.
Cancer Res ; 77(16): 4247-4257, 2017 08 15.
Article En | MEDLINE | ID: mdl-28611045

African American (AA) men exhibit a relatively high incidence and mortality due to prostate cancer even after adjustment for socioeconomic factors, but the biological basis for this disparity is unclear. Here, we identify a novel region on chromosome 4p16.3 that is lost selectively in AA prostate cancer. The negative regulator of G-protein signaling RGS12 was defined as the target of 4p16.3 deletions, although it has not been implicated previously as a tumor-suppressor gene. RGS12 transcript levels were relatively reduced in AA prostate cancer, and prostate cancer cell lines showed decreased RGS12 expression relative to benign prostate epithelial cells. Notably, RGS12 exhibited potent tumor-suppressor activity in prostate cancer and prostate epithelial cell lines in vitro and in vivo We found that RGS12 expression correlated negatively with the oncogene MNX1 and regulated its expression in vitro and in vivo Further, MNX1 was regulated by AKT activity, and RGS12 expression decreased total and activated AKT levels. Our findings identify RGS12 as a candidate tumor-suppressor gene in AA prostate cancer, which acts by decreasing expression of AKT and MNX1, establishing a novel oncogenic axis in this disparate disease setting. Cancer Res; 77(16); 4247-57. ©2017 AACR.


Black or African American/genetics , Genes, Tumor Suppressor , Homeodomain Proteins/biosynthesis , Oncogene Protein v-akt/biosynthesis , Prostatic Neoplasms/genetics , RGS Proteins/genetics , Transcription Factors/biosynthesis , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Heterografts , Homeodomain Proteins/genetics , Humans , Male , Mice , Oncogene Protein v-akt/genetics , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Transfection
12.
Tumour Biol ; 39(6): 1010428317705768, 2017 Jun.
Article En | MEDLINE | ID: mdl-28631564

Signal transducer and activator of transcription 5 and Akt pathways, implicated in signaling transduction downstream of BCR-ABL, play critical roles in the pathogenesis of chronic myeloid leukemia. Therefore, idenication of novel compounds that modulate the activity of such pathways could be a new approach in the treatment of chronic myeloid leukemia. Previous studies have demonstrated that indole-3-carbinol inhibits the proliferation and induces apoptosis of various tumor cells. However, its anticancer activity against chronic myeloid leukemia cells and the underlying mechanism remain unclear. Our data revealed that indole-3-carbinol promoted mitochondrial apoptosis of chronic myeloid leukemia-derived K562 cells, as evidenced by the activation of caspases and poly (ADP-ribose) polymerase cleavage. Treatment with indole-3-carbinol was found to be associated with a decrease in the cellular levels of phospho-Akt and phospho-signal transducer and activator of transcription 5. In addition, real-time polymerase chain reaction analysis showed that the downregulation of genes is regulated by Akt and signal transducer and activator of transcription 5. We also found that treatment with indole-3-carbinol resulted in the activation of the p38 mitogen-activated protein kinase and reduced expression of human telomerase and c-Myc. Collectively, these results demonstrate that the oncogenic signal transducer and activator of transcription 5/Akt pathway is a cellular target for indole-3-carbinol in chronic myeloid leukemia cells. Thus, this clinically tested natural compound can be a potential candidate in the treatment of chronic myeloid leukemia following confirmation with clinical studies.


Indoles/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Oncogene Protein v-akt/genetics , STAT5 Transcription Factor/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Oncogene Protein v-akt/biosynthesis , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , STAT5 Transcription Factor/biosynthesis , Signal Transduction/drug effects , Telomerase/biosynthesis , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/genetics
13.
Tumour Biol ; 39(5): 1010428317698369, 2017 May.
Article En | MEDLINE | ID: mdl-28459364

The influence of celastrol alone or in combination with paclitaxel on survival of anaplastic thyroid carcinoma cells was investigated. In 8505C and SW1736 cells, after treatment of celastrol, cell viability decreased, and cytotoxic activity increased. The protein levels of heat shock protein (hsp) 90, hsp70, Bax, death receptor 5, cleaved caspase-3, cleaved poly (ADP-ribose) polymerase, phospho-extracellular signal-regulated kinase 1/2 (ERK1/2), and phospho-c-Jun N-terminal kinase (JNK) were elevated, and those of Bcl2, phospho-nuclear factor-kappaB (NF-κB), and total and phospho-Akt were reduced. The endoplasmic reticulum stress markers expression and reactive oxygen species production were enhanced. In celastrol-treated cells, N-acetylcysteine increased cell viability and phospho-NF-κB protein levels, and decreased reactive oxygen species production and cytotoxic activity. The protein levels of cyclooxygenase 2, phospho-ERK1/2, phospho-JNK and Bip were diminished. After treatment of both celastrol and paclitaxel, compared with paclitaxel alone, cell viability and the percentage of viable cells were reduced, and death rate and cytotoxic activity were elevated. The protein levels of phospho-ERK1/2, phospho-JNK, Bip, and cyclooxygenase 2, and reactive oxygen species production were enhanced. All of the Combination Index values calculated by Chou-Talalay equation were lower than 1.0, implying the synergism between celastrol and paclitaxel in induction of cell death. In conclusion, our results suggest that celastrol induces cytotoxicity through involvement of Bcl2 family proteins and death receptor, and modulation of phospho-NF-κB, Akt, and mitogen-activated protein kinase in association with endoplasmic reticulum stress and reactive oxygen species production in anaplastic thyroid carcinoma cells. Moreover, celastrol synergizes with paclitaxel in induction of cytotoxicity in anaplastic thyroid carcinoma cells.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasm Proteins/biosynthesis , Paclitaxel/administration & dosage , Thyroid Carcinoma, Anaplastic/drug therapy , Triterpenes/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , NF-kappa B/biosynthesis , NF-kappa B/genetics , Oncogene Protein v-akt/biosynthesis , Oncogene Protein v-akt/genetics , Pentacyclic Triterpenes , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species/metabolism , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology
14.
Biomed Res Int ; 2017: 1243515, 2017.
Article En | MEDLINE | ID: mdl-28386546

Objective. To investigate the inhibitory effect of ethyl acetate extracts of Impatiens balsamina L. on prostate cancer cells. Methods. Impatiens balsamina L. was extracted to get water, ethanol, oil ether, ethyl acetate, and butanol extracts. CCK-8 assay was used to detect the inhibitory effect. Apoptosis rates and cell cycle distribution were detected by flow cytometry. Transwell assay was performed to test the ability of migration. The expressions of Bcl-2, Bax, cleaved-caspase-3, p-ERK, ERK, p-AKT, AKT, cyclin D1, cyclin E, and MMP2 were detected by Western blot. Results. Ethyl acetate extracts had the strongest inhibitory effect. After being treated with different concentrations of ethyl acetate extracts, the percentage of G0/G1 phase increased significantly, cyclin D1 and cyclin E expression decreased, apoptosis rate was significantly higher, and the ability of migration of PC-3 and RV1 was inhibited significantly. Western blot showed that the expressions of Bcl-2, p-ERK, and p-AKT were significantly decreased, but the expressions of Bax and caspase-3 cleavage were increased. Conclusions. Impatiens balsamina L. inhibited the proliferation of human prostate cancer cells; ethyl acetate extracts have the strongest effect. It could inhibit cell proliferation and migration, cause G1 phase arrest, and induce apoptosis probably through inhibition of the AKT and ERK pathways.


Apoptosis/drug effects , Cell Proliferation/drug effects , Drugs, Chinese Herbal/administration & dosage , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Drugs, Chinese Herbal/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Neoplasm Proteins/biosynthesis , Oncogene Protein v-akt/biosynthesis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
15.
Tumour Biol ; 39(3): 1010428317694298, 2017 Mar.
Article En | MEDLINE | ID: mdl-28351297

Eph receptor A4 (EphA4), a member of the erythropoietin-producing hepatocellular (Eph) family, has been reported to upregulate in several tumors. However, the role of EphA4 in multiple myeloma has not been clarified yet. In this study, we found that EphA4 promoted proliferation of multiple myeloma cells via the regulation of cell cycle. Besides, EphA4 was closely related to cell adhesion of multiple myeloma cells and promoted cell adhesion-mediated drug resistance by enhancing the phosphorylation levels of Akt (p-AKT) expression in multiple myeloma. More interestingly, we discovered that EphA4 can interact with cyclin-dependent kinase 5 (CDK5) and regulate its expression in multiple myeloma. CDK5 has been reported to be overexpressed in multiple myeloma which mediated bortezomib resistance and also participated in AKT pathway. And we have also proved the fact. So, we supposed that EphA4 interacted with CDK5 and promoted its expression which in turn enhanced p-AKT expression and promoted cell adhesion-mediated drug resistance in multiple myeloma. Therefore, this study clarifies the molecular mechanism of cell adhesion-mediated drug resistance and may be useful in identifying potential target for treatment of multiple myeloma.


Cyclin-Dependent Kinase 5/biosynthesis , Multiple Myeloma/genetics , Oncogene Protein v-akt/genetics , Receptor, EphA4/genetics , Bortezomib/administration & dosage , Cell Adhesion/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 5/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/pathology , Oncogene Protein v-akt/biosynthesis , Phosphorylation , Receptor, EphA4/biosynthesis , Signal Transduction/drug effects
16.
J Recept Signal Transduct Res ; 37(2): 133-140, 2017 Apr.
Article En | MEDLINE | ID: mdl-27400655

CONTEXT: The histamine H4 receptor functionally expressed on human mast cells and their signaling pathways for the production of IL-13 and RANTES have never been analyzed side by side in a directly comparable manner. OBJECTIVE: Therefore, the aim of the study was to investigate signaling transduction pathways of H4R via ERK1/2, Akt and NFκB leading to the induction of inflammatory cytokine expression. MATERIALS AND METHODS: In the present study, HMC-1 cells and CBMCs were pretreated individually with H4R antagonist JNJ7777120, H1R antagonist mepyramine and signaling molecule inhibitors PD 98059, LY294002, Bay 117082 followed by stimulation was done with or without histamine or 4-MH. Furthermore, the siRNA mediated H4R gene silencing effects are studied at the H4R protein expression level and also signal transduction level. RESULTS: We found that the pretreatment with JNJ7777120 and H4R gene silencing decreased histamine, 4-MH induced phosphorylation of ERK1/2, Akt and NFκB-p65. Moreover, PD 98059, LY294002 and Bay 117082, which respectively inhibited the histamine and 4-methylhistamine induced phosphorylation of ERK1/2, Akt and NFκB-p65 respectively. We also found that the activation of H4R caused the release of IL-13 and RANTES on human mast cells. The MEK inhibitor PD98059 blocked H4R mediated RANTES/CCL5 production by 20.33 pg/ml and inhibited IL-13 generation by 95.71 pg/ml. In contrast, PI3 kinase inhibitor LY294002 had no effect on 4-MH induced RANTES/CCL5 production but blocked IL-13 generation by 117.58 pg/ml. DISCUSSION AND CONCLUSION: These data demonstrate that the H4R activates divergent signaling pathways to induce cytokine and chemokine production in human mast cells.


Chemokine CCL5/genetics , Histamine/metabolism , Interleukin-13/genetics , Mast Cells/metabolism , Receptors, Histamine/metabolism , Chemokine CCL5/biosynthesis , Chromones/administration & dosage , Flavonoids/administration & dosage , Gene Expression Regulation/drug effects , Histamine Antagonists/administration & dosage , Humans , Interleukin-13/biosynthesis , MAP Kinase Signaling System/drug effects , Methylhistamines/administration & dosage , Morpholines/administration & dosage , NF-kappa B/biosynthesis , Oncogene Protein v-akt/biosynthesis , Phosphorylation , Receptors, Histamine/genetics
17.
J Recept Signal Transduct Res ; 37(2): 181-188, 2017 Apr.
Article En | MEDLINE | ID: mdl-27401115

CONTEXT/OBJECTIVE: Cell lines used to study the role of the G protein-coupled receptor 30 (GPR30) or G protein-coupled estrogen receptor (GPER) as a mediator of estrogen responses have yielded conflicting results. This work identified a simple assay to predict cell line competence for pharmacological studies of GPR30. MATERIALS AND METHODS: The phosphorylation or expression levels of ERK1/2, Akt, c-Fos and eNOS were evaluated to assess GPR30 activation in response to known agonists (17ß-estradiol and G-1) in MCF-7 and T-47D breast cancer cell lines and in bovine aortic endothelial cells. GPR30 expression was analyzed by qRT-PCR and Western blot with two distinct antibodies directed at its carboxy and amino terminals. RESULTS: None of the agonists, at any of the concentrations tested, activated any of those target proteins. Additional experiments excluded the disruption of the signaling pathway, interference of phenol red in the culture medium and constitutive proteasome degradation of GPR30 as possible causes for the lack of response of the three cell lines. Analysis of receptor expression showed the absence of clearly detectable GPR30 species of 44 and 50-55 kDa previously identified in cell lines that respond to 17ß-estradiol and G-1. DISCUSSION AND CONCLUSION: Cells that do not express the 44 and 50-55 kDa species do not respond to GPR30 agonists. Thus, the presence or absence of these GPR30 species is a simple and rapid manner to determine whether a given cell line is suitable for pharmacological or molecular studies of GPR30 modulation.


Breast Neoplasms/drug therapy , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Receptors, Estrogen/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Animals , Aorta/cytology , Aorta/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cattle , Cyclopentanes/administration & dosage , Endothelial Cells/cytology , Endothelial Cells/drug effects , Estradiol/administration & dosage , Estrogen Receptor alpha/genetics , Estrogens/agonists , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Oncogene Protein v-akt/biosynthesis , Phosphorylation , Proto-Oncogene Proteins c-fos/biosynthesis , Quinolines/administration & dosage , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects
18.
Int J Oncol ; 50(1): 185-192, 2017 Jan.
Article En | MEDLINE | ID: mdl-27922686

A major goal of breast cancer research is to prevent the molecular events that lead to tumour metastasis. It is well-established that both cytoplasmic and mitochondrial reactive oxygen species (ROS) play important roles in cell migration and metastasis. Accordingly, this study examined the molecular mechanisms of the anti-metastatic effects of NecroX-5, a mitochondrial ROS scavenger. NecroX-5 inhibited lung cancer metastasis by ameliorating migration in a mouse model. In human cancer cells, the inhibition of migration by NecroX-5 is cell type-dependent. We observed that the effect of NecroX-5 correlated with a reduction in mitochondrial ROS, but mitochondrial ROS reduction by MitoQ did not inhibit cell migration. NecroX-5 decreased intracellular calcium concentration by blocking Ca2+ influx, which mediated the inhibition of cell migration, AKT downregulation and the reduction of mitochondrial ROS levels. However, the reduction of mitochondrial ROS was not associated with supressed migration and AKT downregulation. Our study demonstrates the potential of NecroX-5 as an inhibitor of breast cancer metastasis.


Breast Neoplasms/drug therapy , Cell Movement/drug effects , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Oncogene Protein v-akt/biosynthesis , Sulfones/administration & dosage , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Metastasis , Oncogene Protein v-akt/genetics , Organophosphorus Compounds/administration & dosage , Reactive Oxygen Species/metabolism , Ubiquinone/administration & dosage , Ubiquinone/analogs & derivatives , Xenograft Model Antitumor Assays
19.
Actas Urol Esp ; 41(3): 172-180, 2017 Apr.
Article En, Es | MEDLINE | ID: mdl-27726892

OBJECTIVE: To determine the differential protein expression of biomarkers FGFR3, PI3K (subunits PI3Kp110α, PI3KClassIII, PI3Kp85), AKT, p21Waf1/Cip1 and cyclins D1 and D3 in T1 bladder cancer versus healthy tissue and to study their potential role as early recurrence markers. MATERIAL AND METHOD: This is a prospective study that employed a total of 67 tissue samples (55 cases of T1 bladder tumours that underwent transurethral resection and 12 cases of adjacent healthy mucosa). The protein expression levels were assessed using Western blot, and the means and percentages were compared using Student's t-test and the chi-squared test. The survival analysis was conducted using the Kaplan-Meier method and the log-rank test. RESULTS: Greater protein expression was detected for FGFR3, PI3Kp110α, PI3KClassIII, cyclins D1 and D3 and p21Waf1/Cip1 in the tumour tissue than in the healthy mucosa. However, these differences were not significant for PI3Kp85 and AKT. We observed statistically significant correlations between early recurrence and PI3Kp110α, PI3KClassIII, PI3Kp85 and AKT (P=.003, P=.045, P=.050 and P=.028, respectively), between the tumour type (primary vs. recurrence) and cyclin D3 (P=.001), between the tumour size and FGFR3 (P=.035) and between multifocality and cyclin D1 (P=.039). The survival analysis selected FGFR3 (P=.024), PI3Kp110α (P=.014), PI3KClassIII (P=.042) and AKT (P=.008) as markers of early-recurrence-free survival. CONCLUSIONS: There is an increase in protein expression levels in bladder tumour tissue. The overexpression of FGFR3, PI3Kp110α, PI3KClassIII and AKT is associated with increased early-recurrence-free survival for patients with T1 bladder tumours.


Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality , Aged , Aged, 80 and over , Cyclin D1/biosynthesis , Cyclin D2/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Female , Humans , Male , Middle Aged , Neoplasm Staging , Oncogene Protein v-akt/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis , Prognosis , Prospective Studies , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Survival Analysis , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery
20.
J Neurochem ; 140(1): 96-113, 2017 01.
Article En | MEDLINE | ID: mdl-27787894

The astrocyte marker, glial fibrillary acidic protein (GFAP), has essential functions in the brain, but may trigger astroglial scarring when expressed in excess. Docosahexaenoic acid (DHA) is an n-3 fatty acid that is protective during brain development. However, the effect of DHA on GFAP levels of developing brain remains unexplored. Here, we detected that treating developing rats with DHA-enriched fish-oil caused dose-dependent GFAP augmentation. We investigated the mechanism promoting GFAP, hypothesizing the participation of fatty acid-binding protein-7 (FABP7), known to bind DHA. We identified that DHA stimulated FABP7 expression in astrocytes, and FABP7-silencing suppressed DHA-induced GFAP, indicating FABP7-mediated GFAP increase. Further investigation proved FABP7 expression to be phosphatidylinositide 3-kinases (PI3K)/AKT and nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARγ)-dependent. We found that PI3K/AKT activated PPARγ that triggered FABP7 expression via PPARγ-responsive elements within its gene. Towards identifying FABP7-downstream pathways, we considered our previous report that demonstrated cyclin-dependent kinase-5 (CDK5)-PPARγ-protein-protein complex to suppress GFAP. We found that the DHA-induced FABP7 underwent protein-protein interaction with PPARγ, which impeded CDK5-PPARγ formation. Hence, it appeared that enhanced FABP7-PPARγ in lieu of CDK5-PPARγ resulted in increased GFAP. PI3K/AKT not only stimulated formation of FABP7-PPARγ protein-protein complex, but also up-regulated a FABP7-independent MAP-kinase-phosphatase-3 pathway that inactivated CDK5 and hence attenuated CDK5-PPARγ. Overall, our data reveal that via the proximal PI3K/AKT, DHA induces FABP7-PPARγ, through genomic and non-genomic mechanisms, and MAP-kinase-phosphatase-3 that converged at attenuated CDK5-PPARγ and therefore, enhanced GFAP. Accordingly, our study demonstrates a DHA-mediated astroglial hyperactivation, pointing toward a probable injurious role of DHA in brain development.


Astrocytes/metabolism , Docosahexaenoic Acids/pharmacology , Dual Specificity Phosphatase 6/biosynthesis , Fatty Acid-Binding Protein 7/biosynthesis , Glial Fibrillary Acidic Protein/biosynthesis , Oncogene Protein v-akt/biosynthesis , PPAR gamma/biosynthesis , Animals , Astrocytes/drug effects , Brain/drug effects , Brain/growth & development , Brain/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Female , Male , Protein Binding/physiology , Rats , Rats, Wistar , Up-Regulation/drug effects , Up-Regulation/physiology
...