Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 77
1.
Biochem Pharmacol ; 190: 114610, 2021 08.
Article En | MEDLINE | ID: mdl-34010598

Microchromosome maintenance protein 7 (MCM7), a DNA replication permitting factor, plays an essential role in initiating DNA replication. MCM7 is reported to be involved in tumor formation and progression, whereas the expression profile and molecular function of MCM7 in colorectal cancer (CRC) remain unknown. In this study, we aimed to evaluate the clinical significance and biological function of MCM7 in CRC and investigated whether MCM7 can be used for a differential diagnosis in CRC and whether it may serve as a more sensitive proliferation marker for CRC evaluation. Moreover, immunohistochemical analysis of MCM7 was performed in a total of 89 specimens, and high MCM7 expression levels were associated with worse overall survival (OS) in CRC patients. Furthermore, the cell functional test suggested that lentivirus-mediated silencing of MCM7 with shRNA in CRC cells significantly inhibited cellular proliferation and promoted apoptosis in vitro and inhibited tumor growth in vivo. Additionally, mechanistic studies further demonstrated that P21-activated protein kinase 2 (PAK2) was regulated by MCM7 via microarray analysis and cell functional recovery tests, and miR-107 played a role in regulating expression MCM7 via miRNA microarray analysis and 3'UTR reporter assays. Taken together, our results suggest that the miR-107/MCM7/PAK2 pathway may participate in cancer progression and that MCM7 may serve as a prognostic biomarker in CRC.


Apoptosis/physiology , Cell Proliferation/physiology , Colorectal Neoplasms/metabolism , MicroRNAs/biosynthesis , Minichromosome Maintenance Complex Component 7/biosynthesis , p21-Activated Kinases/biosynthesis , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Minichromosome Maintenance Complex Component 7/genetics , Signal Transduction/physiology , Xenograft Model Antitumor Assays/methods , p21-Activated Kinases/genetics
2.
Cell Cycle ; 20(4): 417-433, 2021 02.
Article En | MEDLINE | ID: mdl-33530820

Microglia proliferation is critical for proper development and function of the central nervous system (CNS), while dysregulation of proliferation contributes to pathology. We recently reported that male inducible nitric oxide synthase knockout (iNOS-/-) mice displayed significantly more proliferating microglia in their postnatal cortex than age-matched wildtype (WT) male mice. Moreover, nitric oxide (NO) signaling in mouse microglia greatly upregulates calcium entry through transient receptor potential vanilloid type 2 (TRPV2) channels. Considering that TRPV2 activity restricts astrocytic proliferation within glioma tissues, we investigated the roles of iNOS/NO signaling and TRPV2 expression in the regulation of microglial proliferation in vitro using assays of calcium imaging, immunocytochemistry, western blot, and polymerase chain reaction. Results showed that non-dividing microglia exhibited substantially higher expression of TRPV2 on the plasma membrane and significantly larger calcium influx through TRPV2 channels in comparison to dividing microglia. Additionally, non-dividing WT microglia exhibited significantly more NO production than dividing WT microglia. Furthermore, the NO-donor NOC18 increased the nuclear translocation of nuclear factor of activated T-cells cytoplasmic 2 (NFATC2) and the mRNA of the cyclin-dependent kinase inhibitor p21 and decreased the percentage of dividing WT and iNOS-/- microglia in culture. Importantly, the presence of the TRPV2 inhibitor tranilast abolished these effects of NOC18. Together, results from this study indicated that iNOS/NO signaling inhibits microglial proliferation through TRPV2-mediated calcium influx, nuclear translocation of the transcription factor NFATC2, and p21 expression. [Figure: see text].


Calcium Channels/biosynthesis , Calcium Signaling/physiology , Microglia/metabolism , NFATC Transcription Factors/biosynthesis , Nitric Oxide/biosynthesis , TRPV Cation Channels/biosynthesis , p21-Activated Kinases/biosynthesis , Animals , Calcium Channels/genetics , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/physiology , Cells, Cultured , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/genetics , Nitric Oxide/genetics , TRPV Cation Channels/genetics , Transcription, Genetic/physiology , p21-Activated Kinases/genetics
3.
Oncogene ; 39(20): 4077-4091, 2020 05.
Article En | MEDLINE | ID: mdl-32231273

Muscle-invasive bladder carcinomas (MIBCs) are aggressive genitourinary malignancies. Metastatic urothelial carcinoma of the bladder is generally incurable by current chemotherapy and leads to early mortality. Recent studies have identified molecular subtypes of MIBCs with different sensitivities to frontline therapy, suggesting tumor heterogeneity. We have performed multi-omic profiling of the kinome in bladder cancer patients with the goal of identify therapeutic targets. Our analyses revealed amplification, overexpression, and elevated kinase activity of P21 (RAC1) activated kinase 4 (PAK4) in a subset of Bladder cancer (BLCA). Using bladder cancer cells, we confirmed the role of PAK4 in BLCA cell proliferation and invasion. Furthermore, we observed that a PAK4 inhibitor was effective in curtailing growth of BLCA cells. Transcriptomic analyses identified elevated expression of another kinase, protein tyrosine kinase 6 (PTK6), upon treatment with a PAK4 inhibitor and RNA interference of PAK4. Treatment with a combination of kinase inhibitors (vandetanib and dasatinib) showed enhanced sensitivity compared with either drug alone. Thus, PAK4 may be therapeutically actionable for a subset of MIBC patients with amplified and/or overexpressed PAK4 in their tumors. Our results also indicate that combined inhibition of PAK4 and PTK6 may overcome resistance to PAK4. These observations warrant clinical investigations with selected BLCA patients.


Gene Amplification , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms/enzymology , p21-Activated Kinases/biosynthesis , Cell Line, Tumor , Female , Humans , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , p21-Activated Kinases/genetics
4.
Urol Oncol ; 38(4): 293-304, 2020 04.
Article En | MEDLINE | ID: mdl-31889617

BACKGROUND: Previous studies have shown the prognostic value of PAK1 expression in different tumor patients, including nonmetastatic renal cell carcinoma. In this study, we explored the prognostic and drug predictive value of PAK1 expression in metastatic renal cell carcinoma (mRCC) patients treated with tyrosine kinase inhibitors (TKIs). MATERIALS AND METHODS: We retrospectively enrolled 138 mRCC patients treated with TKIs from a single institution from 2005 to 2014. Analyses were based on 111 patients who met our inclusion criteria. The validation set enrolled 538 RCC patients from The Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma cohort (TCGA KIRC) between 1998 and 2013 in North America. PAK1 expression was assessed by immunohistochemistry (IHC) on tissue microarrays. RESULTS: High PAK1 expression was associated with short overall survival (OS) (P < 0.001) and progression-free survival (PFS) (P = 0.008). Multivariate analyses further indicated that PAK1 expression was an independent prognostic factor for OS (hazard ratio 3.301 [95% confidence interval 2.579-10.899], P < 0.001) and PFS (hazard ratio 3.108 [95% confidence interval 1.795-5.381], P < 0.001). Subgroup analyses suggested that PAK1 was more significant in patients with the intermediate risk group of Heng risk criteria (OS, P = 0.004). Of note, patients treated with Sunitinib showed improved outcome in the low PAK1 subgroup (OS, P = 0.002; PFS, P = 0.013). Finally, relationship was found between PAK1 expression and natural killer cell-mediated cytotoxicity according to gene profile investigation. CONCLUSIONS: High PAK1 expression predicted dismal prognosis in mRCC patients treated with TKIs. Besides, PAK1 was a potential predictor for TKIs treatments.


Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , p21-Activated Kinases/biosynthesis , Adolescent , Adult , Aged , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Female , Humans , Immune Evasion , Kaplan-Meier Estimate , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Retrospective Studies , Young Adult , p21-Activated Kinases/genetics , p21-Activated Kinases/immunology , p21-Activated Kinases/metabolism
5.
Toxicol Lett ; 319: 40-48, 2020 Feb 01.
Article En | MEDLINE | ID: mdl-31706004

Two synthetic tryptamines, namely [3-[2-(diethylamino)ethyl]-1H-indol-4-yl] acetate (4-AcO-DET) and 3-[2-[ethyl(methyl)amino]ethyl]-1H-indol-4-ol (4-HO-MET), are abused by individuals seeking recreational hallucinogens. These new psychoactive substances (NPSs) can cause serious health problems because their adverse effects are mostly unknown. In the present study, we evaluated the cardiotoxicity of 4-AcO-DET and 4-HO-MET using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, electrocardiography (ECG), and the human ether-a-go-go-related gene (hERG) assay. In addition, we analyzed the expression level of p21 (CDC42/RAC)-activated kinase 1 (PAK1), which is known to play various roles in the cardiovascular system. In the MTT assay, 4-AcO-DET- and 4-HO-MET-treated H9c2 cells proliferated in a concentration-dependent manner. Moreover, both substances increased QT intervals (as determined using ECG) in Sprague-Dawley rats and inhibited potassium channels (as verified by the hERG assay) in Chinese hamster ovary cells. However, there was no change in PAK1 expression. Collectively, the results indicated that 4-AcO-DET and 4-HO-MET might cause adverse effects on the cardiovascular system. Further studies are required to confirm the relationship between PAK1 expression and cardiotoxicity. The findings of the present study would provide science-based evidence for scheduling the two NPSs.


Cardiotoxins/toxicity , Hallucinogens/toxicity , Tryptamines/toxicity , Animals , CHO Cells , Cell Line , Cell Survival/drug effects , Cricetulus , ERG1 Potassium Channel/metabolism , Electrocardiography , Male , Myocytes, Cardiac/drug effects , Potassium Channel Blockers/toxicity , Rats , Rats, Sprague-Dawley , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics
6.
Cancer Lett ; 458: 66-75, 2019 08 28.
Article En | MEDLINE | ID: mdl-31121213

Despite the effectiveness of endocrine therapies to treat estrogen receptor-positive (ER+) breast tumours, two thirds of patients will eventually relapse due to de novo or acquired resistance to these agents. Cancer Stem-like Cells (CSCs), a rare cell population within the tumour, accumulate after anti-estrogen treatments and are likely to contribute to their failure. Here we studied the role of p21-activated kinase 4 (PAK4) as a promising target to overcome endocrine resistance and disease progression in ER + breast cancers. PAK4 predicts for resistance to tamoxifen and poor prognosis in 2 independent cohorts of ER + tumours. We observed that PAK4 strongly correlates with CSC activity in metastatic patient-derived samples irrespective of breast cancer subtype. However, PAK4-driven mammosphere-forming CSC activity increases alongside progression only in ER + metastatic samples. PAK4 activity increases in ER + models of acquired resistance to endocrine therapies. Targeting PAK4 with either CRT PAKi, a small molecule inhibitor of PAK4, or with specific siRNAs abrogates CSC activity/self-renewal in clinical samples and endocrine-resistant cells. Together, our findings establish that PAK4 regulates stemness during disease progression and that its inhibition reverses endocrine resistance in ER + breast cancers.


Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , p21-Activated Kinases/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/pharmacology , Female , Fulvestrant/pharmacology , Gene Expression , Humans , MCF-7 Cells , Meta-Analysis as Topic , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prognosis , Small Molecule Libraries/pharmacology , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics
7.
Environ Pollut ; 243(Pt B): 1689-1695, 2018 Dec.
Article En | MEDLINE | ID: mdl-30300874

Aluminum (Al) has neurotoxicity that can result in cognitive dysfunction. Hippocampal dendritic spine loss is a pathological characteristic of cognitive dysfunction. Our previous study reported that Al exposure caused dendritic spine loss in the hippocampus, but the underlying mechanism remains unclear. In this study, rats were orally administered 50, 150 or 450 mg/kg of AlCl3 for 90 days. The dendritic spine density of the CA1 and DG regions was detected by Golgi-Cox staining. The F-actin/G-actin ratio, the expression of drebrin A and the components of the Rac 1/cofilin pathway were measured in the hippocampus. The results obtained showed that AlCl3 caused dendritic spine loss and decreased the F-actin/G-actin ratio. In addition, it was found that AlCl3 downregulated the expression of Rac 1, p-PAK, p-LIMK, p-cofilin and drebrin A and upregulated cofilin expression. Altogether, these results demonstrated that Al inactivated the Rac 1/cofilin pathway by inhibiting the phosphorylation of cofilin and the polymerization of F-actin, resulting in dendritic spine loss in the hippocampus.


Aluminum Chloride/toxicity , Cofilin 1/antagonists & inhibitors , Cognitive Dysfunction/chemically induced , Dendritic Spines/pathology , Hippocampus/pathology , rac1 GTP-Binding Protein/antagonists & inhibitors , Actin Depolymerizing Factors , Actins/analysis , Animals , Cofilin 1/biosynthesis , Lim Kinases/biosynthesis , Male , Neuropeptides/biosynthesis , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , p21-Activated Kinases/biosynthesis , rac1 GTP-Binding Protein/biosynthesis
8.
Proc Natl Acad Sci U S A ; 115(31): 7869-7878, 2018 07 31.
Article En | MEDLINE | ID: mdl-29987050

Endocrine therapy resistance invariably develops in advanced estrogen receptor-positive (ER+) breast cancer, but the underlying mechanisms are largely unknown. We have identified C-terminal SRC kinase (CSK) as a critical node in a previously unappreciated negative feedback loop that limits the efficacy of current ER-targeted therapies. Estrogen directly drives CSK expression in ER+ breast cancer. At low CSK levels, as is the case in patients with ER+ breast cancer resistant to endocrine therapy and with the poorest outcomes, the p21 protein-activated kinase 2 (PAK2) becomes activated and drives estrogen-independent growth. PAK2 overexpression is also associated with endocrine therapy resistance and worse clinical outcome, and the combination of a PAK2 inhibitor with an ER antagonist synergistically suppressed breast tumor growth. Clinical approaches to endocrine therapy-resistant breast cancer must overcome the loss of this estrogen-induced negative feedback loop that normally constrains the growth of ER+ tumors.


Breast Neoplasms/drug therapy , Estrogens/pharmacology , Neoplasm Proteins/biosynthesis , Receptors, Estrogen/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Receptors, Estrogen/genetics , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics , src-Family Kinases/biosynthesis , src-Family Kinases/genetics
9.
Gene Ther ; 25(4): 284-296, 2018 07.
Article En | MEDLINE | ID: mdl-29802374

This study intends to explore the effect of the PAK1 gene silencing on apoptosis and proliferation of hepatocellular carcinoma (HCC) MHCC97-H and HepG2 cells and cells in xenograft tumor. MHCC97-H and HepG2 cells and mice with xenograft tumor in vivo were randomly divided into control, empty vector and PAK1 shRNA groups. Morphology and the expression of green fluorescent protein of MHCC97-H and HepG2 cells and cells in xenograft tumor were observed. MTT assay and flow cytometry were used to detect proliferation, cell cycle and apoptosis of MHCC97-H and HepG2 cells and cells in xenograft tumor. The expressions of PAK1, PCNA, Ki67, Cyclin E, CDK2, p21, p53, Bax and Bcl-2 were measured using the quantitative reverse transcription polymerase chain reaction and western blotting. Compared with the control and empty vector groups, number of adherent cells of MHCC97-H and HepG2 cells and cells in xenograft tumor was reduced, and green fluorescent cells became round and reduced in the PAK1 shRNA group. Cell proliferation, the cells at S phase, the mRNA and protein expressions of PAK1, PCNA, Ki67, Cyclin E, CDK2 and Bcl-2 of MHCC97-H and HepG2 cells and cells in xenograft tumor were decreased, while the cells at G1 phase, apoptosis rate, the mRNA and protein expressions of p21, p53 and Bax of MHCC97-H and HepG2 cells and cells in xenograft tumor were increased in the PAK1 shRNA group. PAK1 gene silencing decreases proliferation of MHCC97-H cells, HepG2 cells and cells in xenograft tumor through the p53/p21 pathway.


Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , p21-Activated Kinases/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Silencing , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Mice , Mice, Nude , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Random Allocation , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/metabolism
10.
Oncotarget ; 7(29): 46042-46055, 2016 Jul 19.
Article En | MEDLINE | ID: mdl-27323857

P21 protein (Cdc42/Rac)-activated kinase 7 (PAK7) can promote neurite outgrowth, induce microtubule stabilization, and activate cell survival signaling pathways. PAK7 expression was found to increase with colon carcinoma progression, but the prognostic value, clinical significance, and underlying mechanisms have not been explored. In my study, the expression of PAK7 was up-related at both the transcriptional and the translational levels in colon tumors compared to that in adjacent normal colon tissue. Patients with PAK7-positive tumors had a lower rate of overall survival (OS) and metastasis-free survival (MFS) (log-rank test, P < 0.001). A Cox proportional hazards model showed that PAK7 expression was an independent prognostic factor for OS (hazard ration [HR], 2.08; 95% confidence interval [CI], 1.16-3.73; P = 0.004) and MFS (HR, 2.88; 95% CI, 1.53-5.42; P < 0.001) in patients with colon cancer. Patients with tumors that were over-expressing PAK7 experienced metastasis, and died within a significantly shorter time after surgery (P < 0.001). Knockdown of PAK7 by a specific short hairpin RNA (shRNA) significantly suppressed the progression of epithelial to mesechymal transition (EMT), migration, and invasion of colon cancer cells in vitro and tumor growth in vivo. However, overexpression of PAK7 significantly promoted these processes. These findings indicate that aberrant PAK7 expression is associated with the occurrence of metastasis and poor clinical outcomes of human colon cancer by promoting the EMT, and the assessment of PAK7 expression might be helpful in predicting metastasis and prognostication for patients with colon cancer.


Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Colonic Neoplasms/pathology , p21-Activated Kinases/biosynthesis , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Disease Progression , Disease-Free Survival , Epithelial-Mesenchymal Transition/physiology , Female , Heterografts , Humans , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Prognosis , Proportional Hazards Models , Young Adult
11.
Cancer Biol Ther ; 17(8): 813-23, 2016 08 02.
Article En | MEDLINE | ID: mdl-27260988

Cancer stem cells (CSC) are tumorigenic and resistant to chemotherapy. In colorectal cancer (CRC), CSCs have been identified by the expression of specific markers, including CD44, Bmi1 and Nanog. Although p21-activated kinase 1 (PAK1), acting downstream of Ras, stimulates Wnt/ß-catenin signaling and is known to play an important role in CRC development and progression, the role of PAK1 in the expression of CSC markers has not previously been investigated. The effect of PAK1 over-expression, knockdown or inhibition on the expression or alteration (in the case of CD44) of CSC markers in human CRC cell lines was measured by immunofluorescence and Western blotting. The effect of PAK1 modulation on tumorigenesis, and on resistance to treatment with 5-fluorouracil (5-FU), was measured by sphere formation in vitro and by growth of xenografted tumors in vivo. The results show that PAK1 activity correlated with the expression of CSC markers and the CD44 isoform profile, and with tumor growth both in vitro and in vivo. Furthermore PAK overexpression partially overcame the inhibition of CRC growth by 5-FU, and PAK inhibition was synergistic with 5-FU treatment. Our findings lay the foundation for a combination therapy in which PAK1 inhibitors targeting CSCs may be combined with conventional 5-FU-based chemotherapy for the treatment of CRC.


Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Fluorouracil/pharmacology , Neoplastic Stem Cells/enzymology , p21-Activated Kinases/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , HCT116 Cells , HT29 Cells , Heterografts , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Up-Regulation , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics
12.
Oncol Res ; 23(3): 119-28, 2016.
Article En | MEDLINE | ID: mdl-26931434

Drug resistance is the leading cause of chemotherapy failure in the treatment of ovarian cancer. So far, little is known about the mechanism of chemoresistance in ovarian cancer. In this study, we explored the mechanism that HSP27 was involved in cisplatin resistance of ovarian cancer both in vitro and clinically. HSP27 protein was found to be upregulated and expressed in cisplatin-resistant ovarian cancer cell line C13*, and HSP27 siRNA transfection reversed the chemoresistance of C13*. We found that HSP27 exerted its chemoresistant role by inhibiting p21 transferring from the nucleus to the plasma through the activation of phosphorylated-Akt pathway. These findings have implications for clinical trials aimed at a potential therapeutic target for ovarian tumors that are refractory to conventional treatment.


Drug Resistance, Neoplasm/genetics , HSP27 Heat-Shock Proteins/genetics , Ovarian Neoplasms/drug therapy , p21-Activated Kinases/biosynthesis , Apoptosis/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cisplatin/administration & dosage , Cytoplasm/genetics , Cytoplasm/metabolism , Female , Gene Knockdown Techniques , HSP27 Heat-Shock Proteins/antagonists & inhibitors , HSP27 Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins , Humans , Molecular Chaperones , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , p21-Activated Kinases/genetics
13.
Oncotarget ; 7(9): 10498-512, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26871290

Diallyl disulfide (DADS) has been shown to have multi-targeted antitumor activities. We have previously discovered that it has a repressive effect on LIM kinase-1 (LIMK1) expression in gastric cancer MGC803 cells. This suggests that DADS may inhibit epithelial-mesenchymal transition (EMT) by downregulating LIMK1, resulting in the inhibition of invasion and growth in gastric cancer. In this study, we reveal that LIMK1 expression is correlated with tumor differentiation, invasion depth, clinical stage, lymph node metastasis, and poor prognosis. DADS downregulated the Rac1-Pak1/Rock1-LIMK1 pathway in MGC803 cells, as shown by decreased p-LIMK1 and p-cofilin1 levels, and suppressed cell migration and invasion. Knockdown and overexpression experiments performed in vitro demonstrated that downregulating LIMK1 with DADS resulted in restrained EMT that was coupled with decreased matrix metalloproteinase-9 (MMP-9) and increased tissue inhibitor of metalloproteinase-3 (TIMP-3) expression. In in vitro and in vivo experiments, the DADS-induced suppression of cell proliferation was enhanced and antagonized by the knockdown and overexpression of LIMK1, respectively. Similar results were observed for DADS-induced changes in the expression of vimentin, CD34, Ki-67, and E-cadherin in xenografted tumors. These results indicate that downregulation of LIMK1 by DADS could explain the inhibition of EMT, invasion and proliferation in gastric cancer cells.


Allyl Compounds/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Disulfides/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lim Kinases/metabolism , Stomach Neoplasms/pathology , Animals , Antigens, CD34/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cofilin 1/metabolism , Down-Regulation , Humans , Ki-67 Antigen/metabolism , Lim Kinases/biosynthesis , Lim Kinases/genetics , Lymphatic Metastasis , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplasm Transplantation , Stomach Neoplasms/mortality , Tissue Inhibitor of Metalloproteinase-3/metabolism , Transplantation, Heterologous , Vimentin/metabolism , p21-Activated Kinases/biosynthesis , rac1 GTP-Binding Protein/biosynthesis , rho-Associated Kinases/biosynthesis
14.
Med Oncol ; 33(3): 22, 2016 Mar.
Article En | MEDLINE | ID: mdl-26820570

Exploring methods for increasing epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) sensitivity has become a major focus in non-small cell lung cancer (NSCLC). Major downstream effectors of the Rho family small guanosine triphosphatases, P21-activated kinases (PAKs) activate the main signaling pathways downstream of EGFR and thus promote tumor cell proliferation. In this study, we explored the expression pattern of phosphorylated PAKs in NSCLC and their potential value as drug targets for treating cancer. The expression and prognostic significance of phosphorylated group I and II PAKs were evaluated in 182 patients with NSCLC. Immunohistochemical analysis revealed low group I PAK expression in normal lung tissues and increased expressed in the cytoplasm, particularly in lung squamous cell carcinoma. Abnormal group I PAK expression was associated with lymph node metastases and high tumor-node-metastases (TNM) stage in NSCLC patients and correlated with poor prognosis. We used group I PAK inhibitor (IPA3) to specifically decrease group I PAK activity in human lung cancer cell lines. Decreased group I PAK activity inhibited cell proliferation and combined IPA3 and EGFR-TKI (gefitinib) treatment inhibited cell proliferation in an obvious manner. Together, our results revealed the PAK expression pattern in NSCLC, and a role for group I PAK in cell proliferation, which provides evidence that decreased PAK activity may have a potential application as a molecular targeted therapy in advanced NSCLC.


Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , p21-Activated Kinases/biosynthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Female , Gefitinib , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Middle Aged , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Quinazolines/therapeutic use , p21-Activated Kinases/genetics
15.
Am J Physiol Renal Physiol ; 310(9): F872-84, 2016 05 01.
Article En | MEDLINE | ID: mdl-26823281

Rats that have recovered from severe proximal tubule (PT) injury induced by uranyl acetate (UA), a toxic stimulus, developed resistance to subsequent UA treatment. We investigated cell cycle status and progression in PT cells in relation to this acquired resistance. Fourteen days after pretreatment with saline (vehicle group) or UA [acute kidney injury (AKI) group], rats were injected with UA or lead acetate (a proliferative stimulus). Cell cycle status (G0/G1/S/G2/M) was analyzed by flow cytometry. The expression of cell cycle markers, cyclin-dependent kinase inhibitors, and phenotypic markers were examined by immunohistochemistry. Cell cycle status in PT cells in the AKI group was comparable to those of the vehicle group. However, more early G1-phase cells (cyclin D1- or Ki67-) and p21+ or p27+ cells were found in the PT of the AKI group than in that of the vehicle group. UA induced G1 arrest and inhibited S phase progression with earlier dedifferentiation and less apoptosis in PT cells of the AKI group. Lead acetate induced proliferation without dedifferentiation but with delayed G0-G1 transition and inhibited S phase progression in PT cells in the AKI group. Sustained p21 and increased p27 expression in PT cells were found in the AKI group in response to UA and lead acetate. PT cells in the AKI group inhibited cell cycle progression by enhanced G1 arrest, probably via p21/p27 modulation as an injury or proliferation response, resulting in cytoresistance to rechallenge injury.


Acute Kidney Injury/pathology , Cell Cycle Checkpoints , Kidney Tubules, Proximal/pathology , Acute Kidney Injury/chemically induced , Animals , Apoptosis/drug effects , Cell Cycle Proteins/biosynthesis , Cell Dedifferentiation/drug effects , Cyclin-Dependent Kinases/biosynthesis , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/drug effects , Male , Organometallic Compounds , Rats , Rats, Sprague-Dawley , Recurrence , p21-Activated Kinases/biosynthesis
16.
Oncogene ; 35(17): 2178-85, 2016 04 28.
Article En | MEDLINE | ID: mdl-26257058

p21-activated kinases (Paks) are Cdc42/Rac-activated serine-threonine protein kinases that regulate several key cancer-relevant signaling pathways, such as the Mek/Erk, PI3K/Akt and Wnt/b-catenin signaling pathways. Pak1 is frequently overexpressed and/or hyperactivated in different human cancers, including human breast, ovary, prostate and brain cancer, due to amplification of the PAK1 gene in an 11q13 amplicon. Genetic or pharmacological inactivation of Pak1 has been shown to reduce proliferation of different cancer cells in vitro and reduce tumor progression in vivo. In this work, we examined the roles of Pak1 in cellular and animal models of PAK1-amplified ovarian cancer. We found that inhibition of Pak1 leads to decreased proliferation and migration in PAK1-amplified/overexpressed ovarian cancer cells, and has no effect in cell that lack such amplification/overexpression. Further, we observed that loss of Pak1 function causes 11q13-amplified ovarian cancer cells to arrest in the G2/M phase of the cell cycle. This arrest correlates with activation of p53 and p21(Cip) and decreased expression of cyclin B1. These findings suggest that small-molecule inhibitors of Pak1 may have a therapeutic role in the ~25% of ovarian cancers characterized by PAK1 gene amplification.


Cell Proliferation/genetics , Ovarian Neoplasms/genetics , p21-Activated Kinases/biosynthesis , Animals , Cell Cycle/genetics , Cell Cycle Checkpoints/genetics , Cyclin B1/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Ovarian Neoplasms/pathology , Signal Transduction , Tumor Suppressor Protein p53/biosynthesis , Xenograft Model Antitumor Assays , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics
17.
Mol Med Rep ; 13(1): 925-32, 2016 Jan.
Article En | MEDLINE | ID: mdl-26647830

Indoleamine 2,3­dioxygenase (IDO), through L­tryptophan depletion, activates general control non­derepressible (GCN) 2 kinase and suppresses T­cell proliferation, in addition to suppressing aerobic glycolysis and glutaminolysis, which are required for these rapidly proliferating cells. A number of, however not all of these alterations, are partially mediated through IDO­induced p53 upregulation. In two­way mixed lymphocyte reactions (MLRs), IDO reduced cellular proliferation. In MLR­derived T­cells, IDO induced the expression levels of p53 and p21, however concurrently reduced the levels of ζ­chain, c­Myc, lactate dehydrogenase A (LDH­A) and glutaminase (GLS)2. However, p53 had no effect on the expression of the above proteins. These results were recapitulated in T­cells activated with anti­CD2, anti­CD3 and anti­CD28 by direct activation of the GCN2 kinase with tryptophanol. In conclusion, IDO, through GCN2 kinase activation, downregulates the levels of TCR­complex ζ­chain and c­Myc, resulting in the suppression of T­cell proliferation and a reduction in the levels of LDH­A and GLS2, which are key enzymes involved in aerobic glycolysis and glutaminolysis, respectively.


Glutaminase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , L-Lactate Dehydrogenase/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , Cell Proliferation/genetics , Glutaminase/genetics , Glycolysis/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , L-Lactate Dehydrogenase/genetics , Mitochondria/genetics , Mitochondria/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/biosynthesis , p21-Activated Kinases/biosynthesis
18.
Front Biosci (Elite Ed) ; 8(1): 205-12, 2016 01 01.
Article En | MEDLINE | ID: mdl-26709656

The vascular endothelium is continuously exposed to cyclic mechanical strain due to the periodic change in vessel diameter as a result of pulsatile blood flow. Since emerging evidence indicates the cyclic strain plays an integral role in regulating endothelial cell function, the present study determined whether application of a physiologic regimen of cyclic strain (6% at 1 hertz) influences the proliferation of human arterial endothelial cells. Prolonged exposure of human dermal microvascular or human aortic endothelial cells to cyclic strain for up to 7 days resulted in a marked decrease in cell growth. The strain-mediated anti-proliferative effect was associated with the arrest of endothelial cells in the G2/M phase of the cell cycle, did not involve cell detachment or cytotoxicity, and was due to the induction of p21. Interestingly, the inhibition in endothelial cell growth was independent of the strain regimen since prolonged application of constant or intermittent 6% strain was also able to block endothelial cell proliferation. The ability of chronic physiologic cyclic strain to inhibit endothelial cell growth represents a previously unrecognized mechanism by which hemodynamic forces maintain these cells in a quiescent, non-proliferative state.


Cell Proliferation , Endothelium, Vascular/cytology , Stress, Mechanical , Cells, Cultured , Endothelium, Vascular/enzymology , Enzyme Induction , Humans , p21-Activated Kinases/biosynthesis
19.
Cell Death Differ ; 23(1): 169-81, 2016 Jan.
Article En | MEDLINE | ID: mdl-26184908

Successful embryo implantation requires functional luminal epithelia to establish uterine receptivity and blastocyst-uterine adhesion. During the configuration of uterine receptivity from prereceptive phase, the luminal epithelium undergoes dynamic membrane reorganization and depolarization. This timely regulated epithelial membrane maturation and precisely maintained epithelial integrity are critical for embryo implantation in both humans and mice. However, it remained largely unexplored with respect to potential signaling cascades governing this functional epithelial transformation prior to implantation. Using multiple genetic and cellular approaches combined with uterine conditional Rac1 deletion mouse model, we demonstrated herein that Rac1, a small GTPase, is spatiotemporally expressed in the periimplantation uterus, and uterine depletion of Rac1 induces premature decrease of epithelial apical-basal polarity and defective junction remodeling, leading to disrupted uterine receptivity and implantation failure. Further investigations identified Pak1-ERM as a downstream signaling cascade upon Rac1 activation in the luminal epithelium necessary for uterine receptivity. In addition, we also demonstrated that Rac1 via P38 MAPK signaling ensures timely epithelial apoptotic death at postimplantation. Besides uncovering a potentially important molecule machinery governing uterine luminal integrity for embryo implantation, our finding has high clinical relevance, because Rac1 is essential for normal endometrial functions in women.


DNA-Binding Proteins/genetics , Embryo Implantation/genetics , Neuropeptides/genetics , Transcription Factors/genetics , p21-Activated Kinases/genetics , p38 Mitogen-Activated Protein Kinases/genetics , rac1 GTP-Binding Protein/genetics , Animals , Blastocyst/metabolism , DNA-Binding Proteins/biosynthesis , Embryo Implantation/physiology , Endometrium/growth & development , Endometrium/metabolism , Epithelium/growth & development , Epithelium/metabolism , Female , Humans , Mice , Neuropeptides/biosynthesis , Signal Transduction/genetics , Transcription Factors/biosynthesis , Uterus/metabolism , Uterus/physiology , p21-Activated Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/biosynthesis , rac1 GTP-Binding Protein/biosynthesis
20.
Mol Cell Biol ; 35(24): 4110-20, 2015 Dec.
Article En | MEDLINE | ID: mdl-26416879

Acetylcholine receptor (AChR) expression in innervated muscle is limited to the synaptic region. Neuron-induced electrical activity participates in this compartmentalization by promoting the repression of AChR expression in the extrasynaptic regions. Here, we show that the corepressor CtBP1 (C-terminal binding protein 1) is present on the myogenin promoter together with repressive histone marks. shRNA-mediated downregulation of CtBP1 expression is sufficient to derepress myogenin and AChR expression in innervated muscle. Upon denervation, CtBP1 is displaced from the myogenin promoter and relocates to the cytoplasm, while repressive histone marks are replaced by activating ones concomitantly to the activation of myogenin expression. We also observed that upon denervation the p21-activated kinase 1 (PAK1) expression is upregulated, suggesting that phosphorylation by PAK1 may be involved in the relocation of CtBP1. Indeed, preventing CtBP1 Ser158 phosphorylation induces CtBP1 accumulation in the nuclei and abrogates the activation of myogenin and AChR expression. Altogether, these findings reveal a molecular mechanism to account for the coordinated control of chromatin modifications and muscle gene expression by presynaptic neurons via a PAK1/CtBP1 pathway.


Alcohol Oxidoreductases/metabolism , DNA-Binding Proteins/metabolism , Muscle Denervation , Muscle, Skeletal/innervation , Myogenin/biosynthesis , p21-Activated Kinases/metabolism , Active Transport, Cell Nucleus/genetics , Alcohol Oxidoreductases/genetics , Animals , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Histones/genetics , Histones/metabolism , Mice , Muscle, Skeletal/metabolism , Myogenin/genetics , Phosphorylation , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Transcriptional Activation/genetics , Up-Regulation , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics
...