Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Neoplasma ; 66(3): 481-486, 2019 May 23.
Article in English | MEDLINE | ID: mdl-30868895

ABSTRACT

Jab1 is a vital subunit of the CSN family and is reported to be overexpressed in numerous cancer types. Due to the importance of Jab1/CSN5 in cancer cell proliferation and survival, Jab1 is considered a promising therapeutic target. Therefore, we evaluated the anticancer effect of the novel Jab1 inhibitor CSN5i-3 in breast cancer cells. In our study, we found that Jab1 was overexpressed in breast cancer tissues and was correlated with poor prognosis in human breast cancer patients. An MTS assay revealed that CSN5i-3 suppressed cell proliferation in the breast cancer cell lines BT474 and SKBR3. We also found that CSN5i-3 significantly induced apoptosis and G1 phase cell cycle arrest in breast cancer cells. A mechanistic investigation revealed that CSN5i-3 inhibited Jab1 expression and increased the level of the apoptosis marker cleaved PARP and the cell-cycle-related protein p27 in BT474 and SKBR3 cells. A nude mouse xenograft model also indicated that CSN5i-3 exerted a potent anticancer effect in vivo. Overall, our study suggested that the Jab1 inhibitor CSN5i-3 might be a promising agent for the treatment of breast cancer in humans and should be studied further.


Subject(s)
Apoptosis , Azepines/pharmacology , Breast Neoplasms , COP9 Signalosome Complex , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins , Peptide Hydrolases , Pyrazoles/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Azepines/therapeutic use , Breast Neoplasms/drug therapy , COP9 Signalosome Complex/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Heterografts , Humans , Imidazoles/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Peptide Hydrolases/metabolism , Pyrazoles/therapeutic use
2.
Aging (Albany NY) ; 11(1): 160-173, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30631005

ABSTRACT

Evidence suggests that altered DNA methylation plays a causative role in the pathogenesis of various cancers, including hepatocellular carcinoma (HCC). Thus, methylated differently expressed genes (MDEGs) could potentially serve as biomarkers and therapeutic targets in HCC. In the present study, screening four genomics profiling datasets (GSE62232, GSE84402, GSE73003 and GSE57956) enabled us to identify a total of 148 MDEGs. A signature was then established based on the top four MDEGs (BRCA1, CAD, CDC20 and RBM8A). Taking clinical variables into consideration, we constructed a risk score system consisting of the four-MDEG signature and the patients' clinical features, which was predictive of prognosis in HCC. The prognostic value of the HCC risk score system was confirmed using TCGA HCC samples. The scores were then used to construct a nomogram, performance of which was evaluated using Harrel's concordance index (C-index) and a calibration curve. The signature-based nomogram for prediction of overall survival in HCC patients exhibited good performance and was superior to traditional staging systems (C-index: 0.676 vs 0.629, P< 0.05). We have thus established a novel risk score system that is predictive of prognosis and is a potentially useful guide for personalized treatment of HCC patients.


Subject(s)
Carcinoma, Hepatocellular/pathology , Gene Expression Profiling , Liver Neoplasms/pathology , RNA, Messenger/metabolism , Aged , Biomarkers, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Methylation , Middle Aged , RNA, Messenger/genetics
3.
Cancer Lett ; 411: 82-89, 2017 12 28.
Article in English | MEDLINE | ID: mdl-28987386

ABSTRACT

Autophagy plays a dual role in many types of cancer, such as hepatocellular carcinoma (HCC). Autophagy seems to be inhibited and functions as a tumor-suppression mechanism in the "inflammation-carcinogenesis" pathway of the liver, including hepatitis B virus and hepatitis C virus, alcoholic steatohepatitis and non-alcoholic steatohepatitis related HCC. However, in established tumors, autophagy plays a tumor-promoting role. Because of the varied function of autophagy in HCC, we hypothesized p62 as a marker to evaluate the autophagic level. Moreover, autophagy is critical in antigen presentation and homeostasis of immune cells and tumor microenvironment. Understanding the intricate relationships of autophagy, inflammation, and immunity provides us with new insights into HCC immunotherapy.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Immunotherapy/methods , Inflammation/pathology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Animals , Autophagy/immunology , Carcinogenesis/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Humans , Inflammation/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology
4.
Oncogene ; 36(8): 1069-1079, 2017 02 23.
Article in English | MEDLINE | ID: mdl-27524414

ABSTRACT

Our previous studies demonstrated that Jab1/Csn5 overexpression is correlated with low survival rates in cancer patients, including nasopharyngeal carcinoma (NPC), breast cancer and hepatocellular carcinoma, and contributes to NPC's resistance to radiotherapy and cisplatin by regulating DNA damage and repair pathways. However, the molecular mechanism by which Jab1/Csn5 expression is upregulated in NPCs has yet to be determined. In the present study, we identified the upstream regulator of Jab1/Csn5 expression and demonstrated its role in intrinsic resistance of NPC cells to treatment with cisplatin. Signal transducer and activator of transcription-3 (Stat3) expression correlates with and contributes to Jab1/Csn5 transcription. Consistently, silencing of Stat3 in tumors reduced Jab1/Csn5 expression, thereby sensitizing NPC cells to cisplatin-induced apoptosis both in vitro and in vivo. Mechanistically, Stat3 transcriptionally regulated Jab1/Csn5. Furthermore, high mRNA expression levels of Stat3 or Jab1 in colon cancer, breast cancer and glioblastoma are associated with significantly shorter survival times from the R2 online database. These findings identify a novel Stat3-Jab1/Csn5 signaling axis in cancer pathogenesis with therapeutic and prognostic relevance.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/genetics , Nasopharyngeal Neoplasms/pathology , Peptide Hydrolases/genetics , STAT3 Transcription Factor/metabolism , Adolescent , Adult , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , COP9 Signalosome Complex , Carcinoma/genetics , Carcinoma/metabolism , Cell Proliferation , Disease Progression , Female , Follow-Up Studies , Humans , Inflammation/genetics , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Staging , Peptide Hydrolases/metabolism , Prognosis , STAT3 Transcription Factor/genetics , Signal Transduction , Transcriptional Activation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
5.
Oncogene ; 35(47): 6096-6108, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27157611

ABSTRACT

Radiotherapy is the standard therapy for nasopharyngeal carcinoma (NPC); however, radioresistance can hinder successful treatment. Here we report that microRNA (miR)-24 acts as a tumor suppressor and radiosensitizer in NPC cells and xenografts by targeting Jab1/CSN5. Although accumulating evidence has shown that Jab1/CSN5 functions as an oncoprotein in human cancers, its regulation through miRs has not been described. In this study, we found that Jab1/CSN5 functioned in a manner opposite to that of miR-24 in NPC tumorigenesis and radioresistance. We demonstrated that miR-24 inhibits Jab1/CSN5 translation via direct binding to its 3' untranslated region (3'UTR) and 5'UTR, leading to tumor growth inhibition, and sensitizes NPC tumors to radiation in vivo. Furthermore, silencing Jab1/CSN5 phenocopied the function of miR-24 in NPC cells after ionizing radiation treatment, resulting in increased apoptosis. Finally, we analyzed 50 paired samples of primary and matched recurrent NPC tissues from 25 NPC patients and subjected them to high-throughput genomic quantitative nuclease protection assay for quantifying simultaneously miR and mRNA levels. Our results showed that miR-24 levels were significantly decreased in recurrent NPC and that levels of Jab1/CSN5, as its target, were higher than those in primary NPC. Together, our findings indicate that miR-24 inhibits NPC tumor growth and increases NPC radiosensitivity by directly regulating Jab1/CSN5 and that both miR-24 and Jab1/CSN5 can serve as prognostic markers for NPC recurrence; this, in turn, may provide a promising therapeutic strategy for reversing NPC radioresistance.


Subject(s)
3' Untranslated Regions , 5' Untranslated Regions , Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Peptide Hydrolases/genetics , RNA Interference , Radiation Tolerance/genetics , Animals , Binding Sites , COP9 Signalosome Complex , Carcinoma/pathology , Carcinoma/radiotherapy , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Silencing , Humans , Mice , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/radiotherapy , Nucleic Acid Conformation , Radiation, Ionizing , Recurrence , Tumor Burden , Xenograft Model Antitumor Assays
6.
Leukemia ; 29(11): 2162-72, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25987255

ABSTRACT

Anaplastic lymphoma kinase-positive (ALK+) anaplastic large-cell lymphoma (ALCL) is an aggressive T-cell non-Hodgkin lymphoma characterized by the t(2;5), resulting in the overexpression of nucleophosmin (NPM)-ALK, which is known to activate the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, resulting in cell cycle and apoptosis deregulation. ALK+ ALCL is also characterized by strong activator protein-1 (AP-1) activity and overexpression of two AP-1 transcription factors, CJUN and JUNB. Here, we hypothesized that a biologic link between AP-1 and AKT kinase may exist, thus contributing to ALCL oncogenesis. We show that JUNB and CJUN bind directly to the AKT1 promoter, inducing AKT1 transcription in ALK+ ALCL. Knockdown of JUNB and CJUN in ALK+ ALCL cell lines downregulated AKT1 mRNA and promoter activity and was associated with lower AKT1 protein expression and activation. We provide evidence that this is a transcriptional control mechanism shared by other cell types even though it may operate in a way that is cell context-specific. In addition, STAT3 (signal transducer and activator of transcription 3)-induced control of AKT1 transcription was functional in ALK+ ALCL and blocking of STAT3 and AP-1 signaling synergistically affected cell proliferation and colony formation. Our findings uncover a novel transcriptional crosstalk mechanism that links AP-1 and AKT kinase, which coordinate uncontrolled cell proliferation and survival in ALK+ ALCL.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, Large-Cell, Anaplastic/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-jun/physiology , Receptor Protein-Tyrosine Kinases/analysis , Transcription Factors/physiology , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Humans , Promoter Regions, Genetic , STAT3 Transcription Factor/physiology , Transcription Factor AP-1/physiology
7.
Oncogene ; 32(22): 2756-66, 2013 May 30.
Article in English | MEDLINE | ID: mdl-22797071

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignancy most common in East Asia and Africa. Radiotherapy and cisplatin-based chemotherapy are the main treatment options. Unfortunately, disease response to concurrent chemoradiotherapy varies among patients with NPC, and many cases are resistant to cisplatin. Increased DNA damage repair is one of the mechanisms contributing to this resistance. Jab1/CSN5 is a multifunctional protein that participates in controlling cell proliferation and the stability of multiple proteins. Jab1 overexpression has been found to correlate with poor prognosis in several tumor types. However, the biological significance of Jab1 activity in response to cancer treatment is unclear. In this study, we used three NPC cell lines (CNE1, CNE2 and HONE1) to investigate the hypothesis that Jab1 positively regulates the DNA repair protein Rad51 and, in turn, cellular response to treatment with DNA-damaging agents such as cisplatin, ionizing radiation (IR) and ultraviolet (UV) radiation. We found that Jab1 was overexpressed in two relatively cisplatin-, IR- and UV-resistant NPC cell lines, and knocking down its expression conferred sensitivity to cisplatin, IR and UV radiation. By contrast, exogenous Jab1 expression enhanced the resistance of NPC cells to cisplatin, IR and UV radiation. Moreover, we provide a mechanism by which Jab1 positively regulated Rad51 through p53-dependent pathway, and increased ectopic expression of Rad51 conferred cellular resistance to cisplatin, IR and UV radiation in Jab1-deficient cells. Taken together, our findings suggest that Jab1 has an important role in the cellular response to cisplatin and irradiation by regulating DNA damage and repair pathways. Therefore, Jab1 is a novel biomarker for predicting the outcome of patients with NPC who are treated with DNA-damaging agents.


Subject(s)
Drug Resistance, Neoplasm/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Nasopharyngeal Neoplasms , Peptide Hydrolases/metabolism , Rad51 Recombinase/metabolism , Radiation Tolerance/genetics , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , COP9 Signalosome Complex , Carcinoma , Cell Line, Tumor , Cell Proliferation , Cisplatin/therapeutic use , DNA Damage , DNA Repair/genetics , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/genetics , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy , Peptide Hydrolases/genetics , RNA Interference , RNA, Small Interfering , Rad51 Recombinase/genetics , Radiation, Ionizing
8.
Leukemia ; 25(5): 856-67, 2011 May.
Article in English | MEDLINE | ID: mdl-21394100

ABSTRACT

p53 is frequently wild type (wt) in diffuse large B-cell lymphoma (DLBCL) associated with t(14;18)(q32;q21) that overexpresses BCL2. Nutlin-3a is a small molecule that activates the p53 pathway by disrupting p53-MDM2 interaction. We show that nutlin-3a activates p53 in DLBCL cells associated with t(14;18)(q32;q21), BCL2 overexpression and wt p53, resulting in cell cycle arrest and apoptosis. Nutlin-3a treatment had similar effects on DLBCL cells of activated B-cell phenotype with wt p53. Cell cycle arrest was associated with upregulation of p21. Nutlin-3a-induced apoptosis was accompanied by BAX and PUMA upregulation, BCL-XL downregulation, serine-70 dephosphorylation of BCL2, direct binding of BCL2 by p53, caspase-9 upregulation and caspase-3 cleavage. Cell death was reduced when p53-dependent transactivation activity was inhibited by pifithrin-α (PFT-α), or PFT-µ inhibited direct p53 targeting of mitochondria. Nutlin-3a sensitized activation of the intrinsic apoptotic pathway by BCL2 inhibitors in t(14;18)-positive DLBCL cells with wt p53, and enhanced doxorubicin cytotoxicity against t(14;18)-positive DLBCL cells with wt or mutant p53, the latter in part via p73 upregulation. Nutlin-3a treatment in a xenograft animal lymphoma model inhibited growth of t(14;18)-positive DLBCL tumors, associated with increased apoptosis and decreased proliferation. These data suggest that disruption of the p53-MDM2 interaction by nutlin-3a offers a novel therapeutic approach for DLBCL associated with t(14;18)(q32;q21).


Subject(s)
Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 18/genetics , Imidazoles/pharmacology , Lymphoma, Large B-Cell, Diffuse/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Translocation, Genetic/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Cell Cycle , Cell Proliferation , Disease Models, Animal , Female , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Mice, SCID , Mutation/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
9.
Oncogene ; 29(46): 6125-37, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-20802511

ABSTRACT

Jun activation domain-binding protein 1 (JAB1) is a multifunctional protein that participates in the control of cell proliferation and the stability of multiple proteins. JAB1 overexpression has been implicated in the pathogenesis of human cancer. JAB1 regulates several key proteins and thereby produces varied effects on cell cycle progression, genome stability and cell survival. However, the biological significance of JAB1 activity in these cellular signaling pathways is unclear. Therefore, we developed mice that were deficient in Jab1 and analyzed the null embryos and heterozygous cells. This disruption of Jab1 in mice resulted in early embryonic lethality due to accelerated apoptosis. Loss of Jab1 expression sensitized both mouse primary embryonic fibroblasts and osteosarcoma cells to γ-radiation-induced apoptosis, with an increase in spontaneous DNA damage and homologous recombination (HR) defects, both of which correlated with reduced levels of the DNA repair protein Rad51 and elevated levels of p53. Furthermore, the accumulated p53 directly binds to Rad51 promoter, inhibits its activity and represents a major mechanism underlying the HR repair defect in Jab1-deficient cells. These results indicate that Jab1 is essential for efficient DNA repair and mechanistically link Jab1 to the maintenance of genome integrity and to cell survival.


Subject(s)
DNA Damage , DNA Repair , Intracellular Signaling Peptides and Proteins/physiology , Peptide Hydrolases/physiology , Animals , Apoptosis , Blastocyst/cytology , COP9 Signalosome Complex , Cell Proliferation , Cell Survival , Embryonic Development , Intracellular Signaling Peptides and Proteins/analysis , Mice , Peptide Hydrolases/analysis , Rad51 Recombinase/physiology , Tumor Suppressor Protein p53/physiology
10.
Oncogene ; 25(19): 2785-94, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16407847

ABSTRACT

N-(4-hydroxyphenyl)retinamide (4HPR), a synthetic retinoid effective in cancer chemoprevention and therapy, is thought to act via apoptosis induction resulting from increased reactive oxygen species (ROS) generation. As ROS can activate MAP kinases and protein kinase C (PKC), we examined the role of such enzymes in 4HPR-induced apoptosis in HNSCC UMSCC22B cells. 4HPR increased ROS level within 1 h and induced activation of caspase 3 and PARP cleavage within 24 h. Activation of MKK3/6 and MKK4, JNK, p38 and ERK was detected between 6 and 12 h, increased up to 24 h and preceded apoptosis. 4HPR-induced activation of these kinases was abrogated by the antioxidants BHA and vitamin C. SP600125, a JNK inhibitor, suppressed 4HPR-induced c-Jun phosphorylation, cytochrome c release from mitochondria and apoptosis. Suppression of JNK1 and JNK2 using siRNA decreased, whereas overexpression of wild type-JNK1 enhanced 4HPR-induced apoptosis. PD169316, a p38, inhibitor suppressed phosphorylation of Hsp27 and apoptosis. PD98059, an MEK1/2 inhibitor, also suppressed ERK1/2 activation and apoptosis induced by 4HPR. Likewise, PKC inhibitor GF109203X suppressed ERK and p38 phosphorylation and PARP cleavage. These data indicate that 4HPR-induced apoptosis is triggered by ROS increase, leading to the activation of the mitogen-activated protein serine/threonine kinases JNK, p38, PKC and ERK, and subsequent apoptosis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/enzymology , Fenretinide/pharmacology , Head and Neck Neoplasms/enzymology , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Carcinoma, Squamous Cell/pathology , Caspase 3 , Caspases/metabolism , Cytochromes c/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Head and Neck Neoplasms/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Tumor Cells, Cultured
11.
J Endocrinol ; 176(1): 103-10, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12525254

ABSTRACT

Macrophage migration inhibitory factor (MIF) is an essential regulator of the macrophage responses to endotoxin. MIF also has the ability to override the anti-inflammatory actions of glucocorticoids during an immune response, and is thus an important pro-inflammatory factor. The presence of MIF in cells of the anterior pituitary has been described, and high levels of MIF in other rapidly proliferating tIssues have also been demonstrated. It has been hypothesised that MIF release from these cells is influenced by the hypothalamo-pituitary-adrenal axis, and that ACTH and MIF are released simultaneously to exert counter-regulatory effects on cortisol. However, another intracellular role for MIF has also been suggested as it has been shown that MIF exerts an effect on the inhibitory cell cycle control protein p27 through an interaction with Jab1, a protein implicated in p27 degradation. We studied MIF expression in different normal and adenomatous human pituitary samples using immunohistochemistry and RT-PCR. There was evidence of co-immunoprecipitation of MIF with Jab1, suggesting an interaction of the two proteins. Our results showed that there is increased expression of MIF protein in the nuclei of all pituitary adenomas compared with normal tIssue (P=0.0067), but there was no statistically significant difference in nuclear MIF expression between the different adenoma types. Nuclear MIF expression correlated positively with p27 and its phosphorylated form in normal tIssue (P=0.0028 and P<0.0001); however, this relationship was not seen in the adenoma samples. Cytoplasmic expression of MIF was found to be variable both in normal and adenomatous samples, with no consistent pattern. MIF mRNA was demonstrated to be present in all tumour and normal samples studied. Somatotroph tumours showed higher MIF mRNA expression compared with normal pituitary or other types of adenomas. In conclusion, MIF is expressed in cell nuclei in pituitary adenomas to a greater extent than in normal pituitary tIssue. We speculate that it may play a role in the control of the cell cycle, but whether its higher level in adenomas is a cause or a consequence of the tumorigenic process remains to be clarified.


Subject(s)
Adenoma/chemistry , Macrophage Migration-Inhibitory Factors/analysis , Pituitary Neoplasms/chemistry , Adult , Aged , Cell Nucleus , Female , Humans , Immunohistochemistry/methods , Macrophage Migration-Inhibitory Factors/genetics , Male , Middle Aged , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
12.
Cancer Res ; 61(19): 7204-10, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585756

ABSTRACT

CD26, a M(r) 110,000 surface-bound ectopeptidase with dipeptidyl peptidase IV (DPPIV) activity, has an array of diverse functional properties, with a role in T-cell physiology and the development of certain human cancers. In this study, we report that surface expression of CD26, through its associated DPPIV enzyme activity, enhanced sensitivity of Jurkat T-cell transfectants to G(2)-M arrest induced by the chemotherapeutic drug, doxorubicin. This was associated with disruption of cell cycle-related events, including hyperphosphorylation and inhibition of p34(cdc2) kinase activity, phosphorylation of cdc25C, and alteration in cyclin B1 expression. In addition, we demonstrate that the addition of exogenous soluble DPPIV enhanced sensitivity of lymphoid tumor cell lines to doxorubicin, suggesting a potentially useful clinical role for CD26/DPPIV in the treatment of selected human hematological malignancies.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Dipeptidyl Peptidase 4/biosynthesis , Doxorubicin/pharmacology , G2 Phase/drug effects , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cyclin B/biosynthesis , Cyclin B1 , Dipeptidyl Peptidase 4/metabolism , G2 Phase/physiology , Humans , Jurkat Cells/cytology , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Mitosis/drug effects , Phosphorylation/drug effects , Transfection , cdc25 Phosphatases/metabolism
13.
J Biol Chem ; 276(1): 413-20, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11020383

ABSTRACT

Overexpression of multidrug resistance genes and their encoded P-glycoproteins is a major mechanism for the development of multidrug resistance in cancer cells. The hepatocarcinogen 2-acetylaminofluorene (2-AAF) efficiently activates rat mdr1b expression. However, the underlying mechanisms are largely unknown. In this study, we demonstrated that a NF-kappa B site on the mdr1b promoter was required for this induction. Overexpression of antisense p65 and I kappa B alpha partially abolished the induction. We then delineated the pathway through which 2-AAF activates NF-kappa B. 2-AAF treatment led to the increase of intracellular reactive oxygen species (ROS) which causes activation of IKK kinases, degradation of I kappa B beta (but not I kappa B alpha), and increase in NF-kappa B DNA binding activity. Consistent with the idea that ROS may participate in mdr1b regulation, antioxidant N-acetylcysteine inhibited the induction of mdr1b by 2-AAF. Overproduction of a physiological antioxidant glutathione (GSH) blocked the activation of IKK kinase complex and NF-kappa B DNA binding. Based on these results, we conclude that 2-AAF up-regulates mdr1b through the generation of ROS, activation of IKK kinase, degradation of I kappa B beta, and subsequent activation of NF-kappa B. This is the first report that reveals the specific cis-elements and signaling pathway responsible for the induction of mdr1b by the chemical carcinogen 2-AAF.


Subject(s)
2-Acetylaminofluorene/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , Carcinogens/pharmacology , Gene Expression Regulation/drug effects , I-kappa B Proteins , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Acetylcysteine/pharmacology , Animals , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Flow Cytometry , Genes, Reporter , Hydrogen Peroxide/pharmacology , I-kappa B Kinase , Oxidative Stress/drug effects , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Transfection , Tumor Cells, Cultured , Up-Regulation/drug effects , ATP-Binding Cassette Sub-Family B Member 4
14.
Mol Cell Biol ; 19(3): 1973-80, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022884

ABSTRACT

Jun N-terminal kinases (JNKs) are serine-threonine kinases that play a critical role in the regulation of cell growth and differentiation. We previously observed that JNK activity is suppressed by all-trans-retinoic acid (t-RA), a ligand for retinoic acid nuclear receptors (RARs), in normal human bronchial epithelial cells, which are growth inhibited by t-RA. In this study, we investigated the mechanism by which t-RA inhibits JNK and the possibility that this signaling event is blocked in non-small cell lung cancer (NSCLC) cells. Virtually all NSCLC cell lines are resistant to the growth-inhibitory effects of t-RA, and a subset of them have a transcriptional defect specific to retinoid nuclear receptors. We found that in NSCLC cells expressing functional retinoid receptors, serum-induced JNK phosphorylation and activity were inhibited by t-RA in a bimodal pattern, transiently within 30 min and in a sustained fashion beginning at 12 h. Retinoid receptor transcriptional activation was required for the late, but not the early, suppression of JNK activity. t-RA inhibited serum-induced JNK activity by blocking mitogen-activated protein (MAP) kinase kinase 4-induced signaling events. This effect of t-RA was phosphatase dependent and involved an increase in the expression of the dual-specificity MAP kinase phosphatase 1 (MKP-1). t-RA did not activate MKP-1 expression or inhibit JNK activity in a NSCLC cell line with retinoid receptors that are refractory to ligand-induced transcriptional activation. These findings provide the first evidence that t-RA suppresses JNK activity by inhibiting JNK phosphorylation. Retinoid receptor transcriptional activation was necessary for the sustained inhibition of JNK activity by t-RA, and this signaling event was disrupted in NSCLC cells with retinoid receptors that are refractory to ligand-induced transcriptional activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins , Immediate-Early Proteins/metabolism , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/metabolism , Tretinoin/metabolism , Dual Specificity Phosphatase 1 , Humans , JNK Mitogen-Activated Protein Kinases , Protein Phosphatase 1 , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured
15.
Mol Cell Biol ; 19(1): 751-63, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9858598

ABSTRACT

The JNK pathway modulates AP-1 activity. While in some cells it may have proliferative and protective roles, in neuronal cells it is involved in apoptosis in response to stress or withdrawal of survival signals. To understand how JNK activation leads to apoptosis, we used PC12 cells and primary neuronal cultures. In PC12 cells, deliberate JNK activation is followed by induction of Fas ligand (FasL) expression and apoptosis. JNK activation detected by c-Jun phosphorylation and FasL induction are also observed after removal of either nerve growth factor from differentiated PC12 cells or KCl from primary cerebellar granule neurons (CGCs). Sequestation of FasL by incubation with a Fas-Fc decoy inhibits apoptosis in all three cases. CGCs derived from gld mice (defective in FasL) are less sensitive to apoptosis caused by KCl removal than wild-type neurons. In PC12 cells, protection is also conferred by a c-Jun mutant lacking JNK phosphoacceptor sites and a small molecule inhibitor of p38 mitogen-activated protein kinase and JNK, which inhibits FasL induction. Hence, the JNK-to-c-Jun-to-FasL pathway is an important mediator of stress-induced neuronal apoptosis.


Subject(s)
Apoptosis , MAP Kinase Kinase Kinase 1 , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinases , Neurons/cytology , Proto-Oncogene Proteins c-jun/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Activation , Fas Ligand Protein , Gene Expression , Mice , Nerve Growth Factors/pharmacology , PC12 Cells , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-jun/genetics , Rats , p38 Mitogen-Activated Protein Kinases
16.
J Neurosci ; 18(14): 5124-35, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9651196

ABSTRACT

Transcription factor c-Jun is proposed to control neuronal cell death and survival, but its activation by N-terminal phosphorylation and the underlying activity of the c-Jun N-terminal kinases (JNKs) remain to be elucidated in the adult mammalian brain. We generated a polyclonal antiserum that specifically recognizes c-Jun phosphorylated at its serine 73 (S73) residue after UV irradiation of 3T3 cells. Disruption of the c-jun locus in 3T3 cells abolished this reaction, and retransfection of the human c-jun at the c-jun-/- background restored it. The phospho-c-Jun antiserum was used to visualize N-terminally phosphorylated c-Jun in the adult rat brain with cellular resolution. Prolonged c-Jun S73 phosphorylation was detected in affected neurons up to 5 d after transient occlusion of medial cerebral artery or up to 50 d after transection of central nerve fiber tracts. After cerebral ischemia-reperfusion, phosphorylation of c-Jun was linked with induced expression of Fas-ligand (APO-1, CD95-ligand), whose gene is a putative c-Jun/AP-1 target, and with terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) reactivity, a marker for apoptosis. After nerve fiber transection, however, lasting c-Jun phosphorylation occurred in axotomized neurons negative for Fas-ligand or TUNEL and regardless of degeneration or survival. In contrast to these lasting phosphorylation patterns, transient seizure activity by pentylenetetrazole provoked only a brief c-Jun phosphorylation and JNK activation. In extracts from ischemic or axotomized brain compartments, c-Jun phosphorylation correlated with enhanced long-term JNK activity, and in-gel kinase assays visualized proteins with sizes corresponding to JNK isoforms as the only c-Jun N-terminally phosphorylating enzymes. These results demonstrate that lasting c-Jun S73 phosphorylation and JNK activity are part of neuronal stress response after neurodegenerative disorders in the adult mammalian brain with Fas-ligand as a putative apoptotic effector.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-jun/metabolism , 3T3 Cells , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death/physiology , Cell Survival/physiology , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Male , Mice , Nerve Fibers/physiology , Neurons/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley , Time Factors
17.
Cell Growth Differ ; 8(3): 283-91, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9056670

ABSTRACT

Retinoids, including retinol and retinoic acid derivatives, inhibit the growth of normal human bronchial epithelial (HBE) cells. Using a lung carcinogenesis model consisting of normal, immortalized, and tumorigenic HBE cells, we showed previously that, compared to normal HBE cells, the tumorigenic HBE cell line 11701 is resistant to the growth-inhibitory effects of all-trans-retinoic acid (t-RA). Retinoid receptor function is preserved in tumorigenic 11701 cells, suggesting that other retinoid signaling components are altered. The activator protein 1 (AP-1) complex is a component of the retinoid signaling pathway and has demonstrated importance in cellular growth and differentiation. Therefore, we investigated whether AP-1 is involved in a retinoid signaling defect in tumorigenic 11701 cells and in retinoid-resistant non-small cell lung cancer (NSCLC) cell lines. We found that t-RA treatment inhibited AP-1 transcriptional activity in normal HBE cells but not in tumorigenic 11701 cells nor in the NSCLC cell lines Calu-1, Calu-6, SKMES-1, and ChaGo K1. We sought mechanisms for this retinoid signaling alteration involving AP-1 in tumorigenic 11701 cells. Basal AP-1 transcriptional activity; AP-1 DNA-binding activity; and the mRNA levels of c-fos, the AP-1 coactivator Jun activation domain-binding protein 1, and the retinoid receptor corepressor, the silencing mediator for retinoid and thyroid hormone receptors (SMRT), were lower in tumorigenic 11701 cells than in normal HBE cells. Transient transfection of tumorigenic 11701 cells with c-fos or CREB binding protein, which is a coactivator of AP-1 and retinoid receptors, enhanced basal AP-1 transcriptional activity but did not alter the effects of t-RA on AP-1 transcriptional activity. These findings provide evidence of a retinoid signaling alteration involving AP-1 in tumorigenic 11701 and NSCLC cells. Furthermore, the inhibitory effect of t-RA on AP-1 transcriptional activity was not restored in tumorigenic 11701 cells by transfection of c-fos, silencing mediator for retinoid and thyroid hormone receptors, Jun activation domain-binding protein 1, or CREB-binding protein, suggesting the involvement of other transcriptional coregulators in this retinoid signaling defect.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2 , Repressor Proteins , Retinoids/pharmacology , Saccharomyces cerevisiae Proteins , Blotting, Northern , Cells, Cultured , DNA Replication/physiology , Epithelium/drug effects , Humans , Minor Histocompatibility Antigens , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Replication Protein C , Signal Transduction/drug effects , Transcription, Genetic/physiology , Transfection , Tumor Cells, Cultured
18.
Nature ; 383(6599): 453-7, 1996 Oct 03.
Article in English | MEDLINE | ID: mdl-8837781

ABSTRACT

The Jun proteins are nuclear proteins that combine with Fos proteins to form a gene-regulatory protein, AP-1. They have highly conserved DNA-binding and dimerization domains, resulting in almost identical sequence-recognition properties. Nevertheless, there are many indications that each Jun protein activates a distinct and only partially overlapping set of AP-1 target genes. Using the more variable activation domain of c-Jun as a bait, we identified a protein, JAB1, that interacts with c-Jun and JunD, but not with JunB or v-Jun. As a result, JAB1 selectively potentiates transactivation by only c-Jun or JunD. In vitro, JAB1 specifically stabilizes complexes of c-Jun or JunD with AP-1 sites and does not affect binding of either JunB or v-Jun. The amino-terminal half of JAB1 is very similar to the amino terminal region of Pad1 from fission yeast, which was identified genetically as a coactivator of a subset of AP-1 target genes. JAB1 and Pad1 are also functionally interchangeable. They define a new group of coactivators that increase the specificity of target gene activation by AP-1 proteins.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Schizosaccharomyces pombe Proteins , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , COP9 Signalosome Complex , Cell Line , Gene Expression Regulation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Peptide Hydrolases , Schizosaccharomyces/metabolism , Trans-Activators/chemistry , Transcription, Genetic , Transcriptional Activation
19.
Mol Cell Biol ; 16(1): 219-27, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8524299

ABSTRACT

Thyroid hormone (T3) receptors (T3Rs) are ligand-modulated transcription factors that bind to thyroid hormone response elements (T3REs) and mediate either positive or negative transcriptional regulation of target genes. In addition, in response to ligand binding, T3Rs can interfere with AP-1 activity and thereby inhibit transcription of AP-1-responsive genes. T3Rs were recently shown to form heterodimers with retinoid X receptors (RXRs), leading to increased binding to T3REs in vitro and potentiation of transcriptional responses in vivo. Here we demonstrate that T3R alpha forms stable heterodimers with RXR alpha in living cells. Most important, we describe a new role for RXR alpha in modulating ligand-dependent T3R alpha activity: heterodimerization with RXR alpha greatly increases transcriptional interference with AP-1 activity, augments T3-dependent transcriptional activation, and potentiates the reversal of ligand-independent activation by T3R alpha. In all three cases, the responses occur at substantially lower T3 concentrations when elicited by T3R alpha plus RXR alpha than by T3R alpha alone. In vitro, the binding of T3 decreases the DNA-binding activity of T3R alpha homodimers but does not affect DNA binding by T3R alpha:RXR alpha heterodimers. We provide evidence that increased activities of T3R alpha at lower T3 concentrations are not due to changes in its T3 binding properties. Instead, the altered response could be mediated by either RXR alpha-induced conformational changes, increased stability of heterodimers over homodimers, especially following T3 binding, or both.


Subject(s)
Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA/metabolism , Humans , Ligands , Molecular Sequence Data , Protein Conformation , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/chemistry , Receptors, Thyroid Hormone/genetics , Retinoid X Receptors , Transcription Factor AP-1/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcriptional Activation , Triiodothyronine/metabolism
20.
EMBO J ; 14(23): 5957-64, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-8846788

ABSTRACT

Growth factors induce c-fos transcription by stimulating phosphorylation of transcription factor TCF/Elk-1, which binds to the serum response element (SRE). Under such conditions Elk-1 could be phosphorylated by the mitogen-activated protein kinases (MAPKs) ERK1 and ERK2. However, c-fos transcription and SRE activity are also induced by stimuli, such as UV irradiation and activation of the protein kinase MEKK1, that cause only an insignificant increase in ERK1/2 activity. However, both of these stimuli strongly activate two other MAPKs, JNK1 and JNK2, and stimulate Elk-1 transcriptional activity and phosphorylation. We find that the JNKs are the predominant Elk-1 activation domain kinases in extracts of UV-irradiated cells and that immunopurified JNK1/2 phosphorylate Elk-1 on the same major sites recognized by ERK1/2, that potentiate its transcriptional activity. Finally, we show that UV irradiation, but not serum or phorbol esters, stimulate translocation of JNK1 to the nucleus. As Elk-1 is most likely phosphorylated while bound to the c-fos promoter, these results suggest that UV irradiation and MEKK1 activation stimulate TCF/Elk-1 activity through JNK activation, while growth factors induce c-fos through ERK activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Genes, fos/genetics , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , HeLa Cells , Humans , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 9 , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serum Response Factor , Transcription, Genetic/genetics , Transfection/genetics , Ultraviolet Rays , ets-Domain Protein Elk-1
SELECTION OF CITATIONS
SEARCH DETAIL
...