Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 115: 105194, 2021 10.
Article in English | MEDLINE | ID: mdl-34365059

ABSTRACT

A series of novel N-aryl-1-(4-sulfamoylphenyl)-5-(thiophen-2-yl)-1H-pyrazole-3-carboxamides was synthesized and examined as inhibitors of cytosolic (human) hCA I and hCA II, and cancer-related transmembrane hCA IX and hCA XII isoenzymes. AC2 was the most selective inhibitor towards cancer-related hCA IX while AC8 and AC9 selectively inhibited hCA XII over off-target isoenzymes. Anticancer effects of the compounds were evaluated towards human oral squamous cell carcinoma (OSCC) cell lines, human mesenchymal normal oral cells, breast (MCF7), prostate (PC3), non-small cell lung carcinoma cells (A549), and non-tumoral fetal lung fibroblast cells (MRC5). Compounds moderately showed cytotoxicity towards cancer cell lines. Among others, AC6 showed cell-specific cytotoxic activity and induced apoptosis in a dose-dependent manner without a significant change in the cell cycle distribution of MCF7. These results suggest that pyrazole-3-carboxamides need further molecular modification to increase their anticancer drug candidate potency.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrases/metabolism , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Neoplasms/enzymology
2.
Adv Exp Med Biol ; 1233: 117-151, 2020.
Article in English | MEDLINE | ID: mdl-32274755

ABSTRACT

Proteostasis regulates key cellular processes such as cell proliferation, differentiation, transcription, and apoptosis. The mechanisms by which proteostasis is regulated are crucial and the deterioration of cellular proteostasis has been significantly associated with tumorigenesis since it specifically targets key oncoproteins and tumor suppressors. Prostate cancer (PCa) is the second most common cause of cancer death in men worldwide. Androgens mediate one of the most central signaling pathways in all stages of PCa via the androgen receptor (AR). In addition to their regulation by hormones, PCa cells are also known to be highly secretory and are particularly prone to ER stress as proper ER function is essential. Alterations in various complex signaling pathways and cellular processes including cell cycle control, transcription, DNA repair, apoptosis, cell adhesion, epithelial-mesenchymal transition (EMT), and angiogenesis are critical factors influencing PCa development through key molecular changes mainly by posttranslational modifications in PCa-related proteins, including AR, NKX3.1, PTEN, p53, cyclin D1, and p27. Several ubiquitin ligases like MDM2, Siah2, RNF6, CHIP, and substrate-binding adaptor SPOP; deubiquitinases such as USP7, USP10, USP26, and USP12 are just some of the modifiers involved in the regulation of these key proteins via ubiquitin-proteasome system (UPS). Some ubiquitin-like modifiers, especially SUMOs, have been also closely associated with PCa. On the other hand, the proteotoxicity resulting from misfolded proteins and failure of ER adaptive capacity induce unfolded protein response (UPR) that is an indispensable signaling mechanism for PCa development. Lastly, ER-associated degradation (ERAD) also plays a crucial role in prostate tumorigenesis. In this section, the relationship between prostate cancer and proteostasis will be discussed in terms of UPS, UPR, SUMOylation, ERAD, and autophagy.


Subject(s)
Prostatic Neoplasms/metabolism , Proteostasis , Deubiquitinating Enzymes/metabolism , Endoplasmic Reticulum-Associated Degradation , Humans , Male , Prostatic Neoplasms/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Unfolded Protein Response
3.
J BUON ; 19(4): 992-8, 2014.
Article in English | MEDLINE | ID: mdl-25536607

ABSTRACT

PURPOSE: Ponatinib (P) has been used for the treatment of chronic myeloid leukemia (CML) and it is known that inhibition of BCR-ABL fusion protein by ponatinib induces apoptosis of CML cells. Epigallocatechin-3-gallate (EGCG), which is a polyphenol in green tea, induces apoptosis in different types of cancer cells. The purpose of this study was to determine the cytotoxic and apoptotic effects of ponatinib and EGCG combination in K562 CML cell line. This study also aimed to detect alterations of the expression levels of cell cycle-regulation related genes after ponatinib and EGCG combination in K562 CML cell line. METHODS: The cytotoxic effects of the compounds on K562 cells were determined in a time-and dose-dependent manner by using WST-1 analysis. The combination index (CI) isobologram was used to analyze the data. Apoptotic effects of P-EGCG were defined by flow cytometry and gene expressions were detected by RT-qPCR. RESULTS: IC50values of ponatinib and EGCG were 87.13 nM and 50µM, respectively. CI value of the P-EGCG was 0.658 and the combination showed synergistic effect (ED90 value: 28.39 nM ponatinib, 117.12 µg/ml EGCG). Ponatinib, EGCG and P-EGCG induced apoptosis compared to control cells. CyclinD1 and CDC25A were downregulated by P-EGCG by 2.49 and 2.63-fold, respectively. TGF-ß2 was upregulated by 4.57-fold. CONCLUSION: EGCG possesses cytotoxic and apoptotic properties and may cooperate with the growth inhibiting activity of ponatinib synergistically against CML cells. P-EGCG mediated apoptosis might be associated with upregulation of TGF-ß2 gene and downregulation of cyclinD1 and CDC25A genes.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Catechin/analogs & derivatives , Cell Cycle/genetics , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Pyridazines/pharmacology , Catechin/pharmacology , Fusion Proteins, bcr-abl , Gene Expression/drug effects , Humans , K562 Cells
4.
J Steroid Biochem Mol Biol ; 141: 26-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24434284

ABSTRACT

It has been reported that NKX3.1 an androgen-regulated homeobox gene restricted to prostate and testicular tissues, encodes a homeobox protein, which transcriptionally regulates oxidative damage responses and enhances topoisomerase I re-ligation by a direct interaction with the ATM protein in prostate cells. In this study, we aimed to investigate the role of NKX3.1 in DNA double-strand break (DSB) repair. We demonstrate that the DNA damage induced by CPT-11 (irinotecan, a topo I inhibitor), doxorubicin (a topo II inhibitor), and H2O2 (a mediator of oxidative damage), but not by etoposide (another topo II inhibitor), is negatively influenced by NKX3.1 expression. We also examined γH2AX((S139)) foci formation and observed that the overexpression of NKX3.1 resulted a remarkable decrease in the formation of γH2AX((S139)) foci. Intriguingly, we observed in NKX3.1 silencing studies that the depletion of NKX3.1 correlated with a significant decrease in the levels of p-ATM((S1981)) and γH2AX((S139)). The data imply that the DNA damage response (DDR) can be altered, perhaps via a decrease in the topoisomerase I re-ligation function; this is consistent with the physical association of NKX3.1 with DDR mediators upon treatment of both PC-3 and LNCaP cells with CPT-11. Furthermore, the depletion of NKX3.1 resulted in a G1/S progression via the facilitation of an increase in E2F stabilization concurrent with the suppressed DDR. Thus, the topoisomerase I inhibitor-mediated DNA damage enhanced the physical association of NKX3.1 with γH2AX((S139)) on the chromatin in LNCaP cells, whereas NKX3.1 in the soluble fraction was associated with p-ATM((S1981)) and RAD50 in these cells. Overall, the data suggest that androgens and NKX3.1 expression regulate the progression of the cell cycle and concurrently activate the DDR. Therefore, androgen withdrawal may augment the development of an error-prone phenotype and, subsequently, the loss of DNA damage control during prostate cancer progression.


Subject(s)
DNA Damage , Homeodomain Proteins/physiology , Transcription Factors/physiology , Acid Anhydride Hydrolases , Ataxia Telangiectasia Mutated Proteins/metabolism , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle , Cell Line, Tumor , Cyclin D1/metabolism , DNA Repair , DNA Repair Enzymes/metabolism , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Etoposide/pharmacology , Histones/metabolism , Humans , Hydrogen Peroxide/pharmacology , Irinotecan , Male , Metribolone/pharmacology , Mutagens/pharmacology , Phosphorylation , Prostatic Neoplasms , Protein Processing, Post-Translational , Testosterone Congeners/pharmacology
5.
Tumour Biol ; 35(2): 1531-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24122279

ABSTRACT

Animal and cell culture studies have showed that boron and its derivatives may be promising anticancer agents in prostate cancer treatment. Thus, DU145 cells were treated with disodium pentaborate decahydrate (DPD) for 24, 48, and 72 h in order to investigate the inhibitor effect and mechanisms of DPD. Then, cell proliferation, telomerase enzyme activity, actin polymerization, and apoptosis were detected by WST-1 assay, qRT-PCR, immunofluorescence labeling, and flow cytometry, respectively. We found that DPD inhibited the growth of human prostate cancer cell line DU145 at the concentration of 3.5 mM for 24 h. Our results demonstrated that 7 mM of DPD treatment prevented the telomerase enzyme activity at the rate of 38 %. Furthermore, DPD has an apoptotic effect on DU145 cells which were examined by labeling DNA breaks. With 7 mM of DPD treatment, 8, 14, and 41 % of apoptotic cells were detected for 24, 48, and 72 h, respectively. Additionally, immunofluorescence labeling showed that the normal organization of actin filaments was disrupted in DPD-exposed cells, which is accompanied by the alteration of cell shape and by apoptosis in targeted cells. Taken together, the results indicate that DPD may exert its cytotoxicity at least partly by interfering with the dynamic properties of actin polymerization and decreasing the telomerase activity. Eventually, for the first time, the results of this study showed that DPD suppressed the activity of telomerase in DU145 cells, and therefore, we suggested that DPD could be an important agent for its therapeutic potential in the treatment of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Borates/pharmacology , Prostatic Neoplasms/drug therapy , Telomerase/genetics , Actins/genetics , Actins/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Telomerase/biosynthesis
6.
Mol Cell Endocrinol ; 383(1-2): 38-47, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24325868

ABSTRACT

HOXB13 is a homeobox protein that is expressed in normal adult prostate and colon tissues; however, its deregulated expression was evidenced in various malignancies. To characterize the putative role of HOXB13 in cell cycle progression, we performed overexpression and siRNA-mediated knockdown studies in PC-3 and LNCaP cells. Immunohistochemistry (IHC) analyses were also performed using formalin-fixed, paraffin-embedded tissues containing normal, H-PIN and PCa sections from 20 radical prostatectomy specimens. Furthermore, when the role of HOXB13 during cell cycle progression, association with cyclins, cell growth and colony formation using real-time cell proliferation were assessed, we observed that ectopic expression of HOXB13 accumulated cells at G1 through decreasing the cyclin D1 level by promoting its ubiquitination and degradation. This loss slowed S phase entry in both cell lines examined, with an associated decrease in pRb((S780) and (S795)) phosphorylations. Contrary, siRNA-mediated depletion of HOXB13 expression noticeably increased cyclin levels, stabilized E2F1 and CDC25C, subsequent to increased pRb phosphorylations. This increase in Cyclin B1 and CDC25C both together facilitated activation of cyclin B complex via dephosphorylating CDK1((T14Y15)), and resumed the G2/M transition after nocodazole synchronization. Despite an increase in the total expression level and cytoplasmic retention of HOXB13 in H-PIN and PCa samples that were observed via IHC evaluation of prostate tissues, HOXB13 depletion facilitated to an increase in PC-3 and LNCaP cell proliferation. Thus, we suggest that HOXB13 expression is required for cell cycle regulation, and increases by an unknown mechanism consequent to its functional loss in cancer.


Subject(s)
G1 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Prostatic Neoplasms/genetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Phosphorylation , Prostate/metabolism , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteolysis , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Ubiquitination , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
7.
Mol Carcinog ; 53(2): 85-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-22911881

ABSTRACT

As a link between inflammation and cancer has been reported in many studies, we established an in vitro model of prostatic inflammation to investigate the loss of androgen receptor (AR)-mediated signaling in androgen responsive prostate cell lines. First, the U937 monocyte cell line was differentiated into macrophages using phorbol acetate (PMA), and cells were induced with lipopolysaccharide (LPS) for cytokine secretion. Next, the cytokine levels (TNFα, IL-6, and IL1ß) in conditioned media (CM) were analyzed. Prostate cells were then fed with CM containing varying concentrations of TNFα, and IkB degradation, nuclear factor kappa B (NFκB) translocation and transactivation, and the expression of matrix metalloproteinase-8 (MMP8) and matrix metalloproteinase-9 (MMP9) were then assessed. As a result of CM treatment, ubiquitin-mediated AR degradation, which was restored using anti-TNFα antibody neutralization, led to both a decrease in KLK4, PSA, and NKX3.1 expression levels and the upregulation of GPX2. In addition to the loss of AR, acute and chronic CM exposure resulted in p53 degradation and consequent p21 downregulation, which was also restored by either androgen administration or ectopic NKX3.1 expression via the stabilization of MDM2 levels in LNCaP cells. Additionally, CM treatment enhanced H2AX((S139)) phosphorylation (a hallmark of DNA damage) and genetic heterogeneity in the absence of androgens in prostate cells without activating mitochondrial apoptosis. Thus, the data suggest that inflammatory cytokine exposure results in the loss of AR and p53 signaling in prostate cells and facilitates genetic heterogeneity via ROS accumulation to promote prostate carcinogenesis.


Subject(s)
Androgens/metabolism , Inflammation/metabolism , Prostatitis/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Androgens/genetics , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/genetics , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Kallikreins/genetics , Kallikreins/metabolism , Lysine/analogs & derivatives , Lysine/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatitis/drug therapy , Prostatitis/pathology , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Reactive Oxygen Species/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
8.
Mol Cell Endocrinol ; 350(1): 107-17, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22155408

ABSTRACT

We recently reported that hematological and neurological expressed 1 (HN1) is a ubiquitously expressed, EGF-regulated gene. Expression of HN1 in prostate cell lines down-regulates PI3K-dependent Akt activation. Here, we investigate whether HN1 is regulated by androgens through the putative androgen response elements (AREs) found in its promoter. Knockdown of HN1 expression by siRNA silencing leads to an increase in Akt((S473)) phosphorylation, resulting in the translocation of androgen receptor (AR) to the nucleus; these effects can be abrogated by the non-specific Akt inhibitor LY294002 but not by the ERK inhibitor PD98059. Furthermore, HN1 overexpression correlates with an increase in ubiquitination-mediated degradation (a consequence of the decrease in S213/210 phosphorylation of AR), ultimately resulting in the down-regulation of AR-mediated expression of the KLK3, KLK4, NKX3.1 and STAMP2 genes. We also found that HN1 overexpression suppresses colony formation as well as R1881-mediated growth in LNCaP cells, while it has the opposite effect (increasing colony formation but not proliferation) in PC-3 and DU145 cells. Therefore, we suggest that HN1 maintains a balance between the androgen-regulated nuclear translocation of AR and steady-state Akt phosphorylation, predominantly in the absence of androgens. If so, the balance between cell growth and EGF- and AR-signaling must be tightly regulated by HN1. This work has important implications for prostate cancer research, as AR, EGFR and HN1 are known to be highly expressed in prostate adenocarcinomas.


Subject(s)
Nerve Tissue Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Androgen/metabolism , Transcriptional Activation , Androgens/pharmacology , Androgens/physiology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Dexamethasone/pharmacology , Estradiol/pharmacology , Gene Expression , Gene Expression Regulation , Glucocorticoids/pharmacology , Humans , Kallikreins/genetics , Kallikreins/metabolism , Leupeptins , Male , Metribolone/pharmacology , Microtubule-Associated Proteins , Nerve Tissue Proteins/genetics , Nuclear Proteins , Phosphorylation , Prostatic Neoplasms , Proteasome Inhibitors , Protein Transport , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism
9.
Biochem Biophys Res Commun ; 414(1): 123-8, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21945437

ABSTRACT

NKX3.1 is an androgen-regulated homeobox gene that encodes a tissue-restricted transcription factor, which plays an important role in the differentiation of the prostate epithelium. Thus, the role of NKX3.1 as a functional topoisomerase I activity enhancer in cell cycle regulation and the DNA damage response (DDR) was explored in prostate cancer cell lines. As an early response to DNA damage following CPT-11 treatment, we found that there was an increase in the γH2AX(S139) foci number and that total phosphorylation levels were reduced in PC-3 cells following ectopic NKX3.1 expression as well as in LNCaP cells following androgen administration. Furthermore, upon drug treatment, the increase in ATM(S1981) phosphorylation was reduced in the presence of NKX3.1 expression, whereas DNA-PKcs expression was increased. Additionally, phosphorylation of CHK2(T68) and NBS1(S343) was abrogated by ectopic NKX3.1 expression, compared with the increasing levels in control PC-3 cells in a time-course experiment. Finally, NKX3.1 expression maintained a high cyclin D1 expression level regardless of drug treatment, while total γH2AX(S139) phosphorylation remained depleted in PC-3, as well as in LNCaP, cells. Thus, we suggest that androgen regulated NKX3.1 maintains an active DDR at the intra S progression and contributes to the chemotherapeutic resistance of prostate cancer cells to DNA damaging compounds.


Subject(s)
DNA Damage , Homeodomain Proteins/physiology , Prostatic Neoplasms/pathology , S Phase , Transcription Factors/physiology , Androgens/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Histones/pharmacology , Homeodomain Proteins/genetics , Humans , Irinotecan , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Transcription Factors/genetics
10.
DNA Cell Biol ; 30(6): 419-29, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21323578

ABSTRACT

As the molecular mechanism of ß-catenin deregulation is not well understood, and stabilized ß-catenin is known to translocate into the nucleus and activate genes for proliferation, a novel regulatory factor, hematological and neurological expressed 1 (HN1), for Akt-GSK3ß-ß-catenin axis is reported here. In our studies, HN1 gene structure was characterized. HN1 expression was found to be epidermal growth factor-responsive in PC-3 cells, and protein expression was also upregulated in PC-3 and LNCaP but not in DU145 cells. Additionally, HN1 was found to be downregulated by the specific AKT inhibitor wortmannin but not with PI3K or MAPK inhibitors, LY294002 and PD98059, respectively, in PC-3 and MCF-7 cells. Further, siRNA-mediated knockdown of HN1 resulted in considerable increase in Akt((S473)) and GSK3ß((S9),(Y216)) phosphorylations; moreover, subsequent accumulation of ß-catenin, increase in c-myc expression, and nuclear accumulation of cyclin D1 were observed in PC-3 cells. Knockdown of HN1 also resulted in prolongation of G(1) phase in cell cycle, increasing tetraploidy, presumably because of cells escaping from abnormal mitosis in PC-3 cells. Consistently, overexpression of HN1 reversed the cell-cycle-specific observations, resulted in accumulation of cells in G(2)/M, and reduced the proliferation rate, which were investigated using flow cytometry and methylthiazol tetrazolium assays. As activating mutations of ß-catenin have been demonstrated in late-stage tumors, and ß-catenin stabilization was correlated with poor prognosis in previous reports, epidermal growth factor-upregulated HN1 expression might have a role in deregulating the AKT-GSK3ß((S9))-mediated signaling as a novel compensating mechanism.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Neoplasm Proteins/genetics , Nerve Tissue Proteins/metabolism , Prostate/cytology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Amino Acid Sequence , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Chromones/pharmacology , Cloning, Molecular , Cyclin B1/metabolism , Cyclin D1/metabolism , Down-Regulation , Epidermal Growth Factor/pharmacology , Flavonoids/pharmacology , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Humans , Male , Molecular Sequence Data , Morpholines/pharmacology , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Phosphorylation , Tetraploidy , Up-Regulation , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...