Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Cancer Res ; 13(12): 1544-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26358474

ABSTRACT

UNLABELLED: Signaling via the MET receptor tyrosine kinase has been implicated in crosstalk with cellular responses to DNA damage. Our group previously demonstrated that MET inhibition in tumor cells with deregulated MET activity results in radiosensitization via downregulation of the ATR-CHK1-CDC25 pathway, a major signaling cascade responsible for intra-S and G2-M cell-cycle arrest following DNA damage. Here we aimed at studying the potential therapeutic application of ionizing radiation in combination with a MET inhibitor, EMD-1214063, in p53-deficient cancer cells that harbor impaired G1-S checkpoint regulation upon DNA damage. We hypothesized that upon MET inhibition, p53-deficient cells would bypass both G1-S and G2-M checkpoints, promoting premature mitotic entry with substantial DNA lesions and cell death in a greater extent than p53-proficient cells. Our data suggest that p53-deficient cells are more susceptible to EMD-1214063 and combined treatment with irradiation than wild-type p53 lines as inferred from elevated γH2AX expression and increased cytotoxicity. Furthermore, cell-cycle distribution profiling indicates constantly lower G1 and higher G2-M population as well as higher expression of a mitotic marker p-histone H3 following the dual treatment in p53 knockdown isogenic variant, compared with the parental counterpart. IMPLICATIONS: The concept of MET inhibition-mediated radiosensitization enhanced by p53 deficiency is of high clinical relevance, as p53 is frequently mutated in numerous types of human cancer. The current data point for a therapeutic advantage for an approach combining MET targeting along with DNA-damaging agents for MET-positive/p53-negative tumors.


Subject(s)
Cell Cycle Checkpoints/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridazines/pharmacology , Pyrimidines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53/deficiency , Apoptosis , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans
2.
Cell Death Dis ; 6: e1861, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26313912

ABSTRACT

All-trans retinoic acid (ATRA), a pan-retinoic acid receptor (RAR) agonist, is, along with other retinoids, a promising therapeutic agent for the treatment of a variety of solid tumors. On the one hand, preclinical studies have shown promising anticancer effects of ATRA in breast cancer; on the other hand, resistances occurred. Autophagy is a cellular recycling process that allows the degradation of bulk cellular contents. Tumor cells may take advantage of autophagy to cope with stress caused by anticancer drugs. We therefore wondered if autophagy is activated by ATRA in mammary tumor cells and if modulation of autophagy might be a potential novel treatment strategy. Indeed, ATRA induces autophagic flux in ATRA-sensitive but not in ATRA-resistant human breast cancer cells. Moreover, using different RAR agonists as well as RARα-knockdown breast cancer cells, we demonstrate that autophagy is dependent on RARα activation. Interestingly, inhibition of autophagy in breast cancer cells by either genetic or pharmacological approaches resulted in significantly increased apoptosis under ATRA treatment and attenuated epithelial differentiation. In summary, our findings demonstrate that ATRA-induced autophagy is mediated by RARα in breast cancer cells. Furthermore, inhibition of autophagy results in enhanced apoptosis. This points to a potential novel treatment strategy for a selected group of breast cancer patients where ATRA and autophagy inhibitors are applied simultaneously.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Receptors, Retinoic Acid/agonists , Tretinoin/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Differentiation/drug effects , Cell Line, Tumor , Chloroquine/pharmacology , Drug Resistance, Neoplasm/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Macrolides/pharmacology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
3.
Eur J Histochem ; 59(2): 2481, 2015 May 05.
Article in English | MEDLINE | ID: mdl-26150155

ABSTRACT

Autophagy assures cellular homeostasis, and gains increasing importance in cancer, where it impacts on carcinogenesis, propagation of the malignant phenotype and development of resistance. To date, its tissue-based analysis by immunohistochemistry remains poorly standardized. Here we show the feasibility of specifically and reliably assessing the autophagy markers LC3B and p62 (SQSTM1) in formalin fixed and paraffin embedded human tissue by immunohistochemistry. Preceding functional experiments consisted of depleting LC3B and p62 in H1299 lung cancer cells with subsequent induction of autophagy. Western blot and immunofluorescence validated antibody specificity, knockdown efficiency and autophagy induction prior to fixation in formalin and embedding in paraffin. LC3B and p62 antibodies were validated on formalin fixed and paraffin embedded cell pellets of treated and control cells and finally applied on a tissue microarray with 80 human malignant and non-neoplastic lung and stomach formalin fixed and paraffin embedded tissue samples. Dot-like staining of various degrees was observed in cell pellets and 18/40 (LC3B) and 22/40 (p62) tumors, respectively. Seventeen tumors were double positive for LC3B and p62. P62 displayed additional significant cytoplasmic and nuclear staining of unknown significance. Interobserver-agreement for grading of staining intensities and patterns was substantial to excellent (kappa values 0.60 - 0.83). In summary, we present a specific and reliable IHC staining of LC3B and p62 on formalin fixed and paraffin embedded human tissue. Our presented protocol is designed to aid reliable investigation of dysregulated autophagy in solid tumors and may be used on large tissue collectives.


Subject(s)
Autophagy , Microtubule-Associated Proteins/analysis , RNA-Binding Proteins/analysis , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Feasibility Studies , Fluorescent Antibody Technique , Formaldehyde , Gene Knockdown Techniques , Humans , Immunohistochemistry , Observer Variation , Paraffin Embedding , Reproducibility of Results , Tissue Fixation
4.
Cell Death Dis ; 6: e1778, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26043078

ABSTRACT

While many anticancer therapies aim to target the death of tumor cells, sophisticated resistance mechanisms in the tumor cells prevent cell death induction. In particular enzymes of the glutathion-S-transferase (GST) family represent a well-known detoxification mechanism, which limit the effect of chemotherapeutic drugs in tumor cells. Specifically, GST of the class P1 (GSTP1-1) is overexpressed in colorectal tumor cells and renders them resistant to various drugs. Thus, GSTP1-1 has become an important therapeutic target. We have recently shown that thiazolides, a novel class of anti-infectious drugs, induce apoptosis in colorectal tumor cells in a GSTP1-1-dependent manner, thereby bypassing this GSTP1-1-mediated drug resistance. In this study we investigated in detail the underlying mechanism of thiazolide-induced apoptosis induction in colorectal tumor cells. Thiazolides induce the activation of p38 and Jun kinase, which is required for thiazolide-induced cell death. Activation of these MAP kinases results in increased expression of the pro-apoptotic Bcl-2 homologs Bim and Puma, which inducibly bind and sequester Mcl-1 and Bcl-xL leading to the induction of the mitochondrial apoptosis pathway. Of interest, while an increase in intracellular glutathione levels resulted in increased resistance to cisplatin, it sensitized colorectal tumor cells to thiazolide-induced apoptosis by promoting increased Jun kinase activation and Bim induction. Thus, thiazolides may represent an interesting novel class of anti-tumor agents by specifically targeting tumor resistance mechanisms, such as GSTP1-1.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Glutathione S-Transferase pi/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Thiazoles/pharmacology , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caco-2 Cells , Cell Line , Cisplatin/pharmacology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Enzyme Activation , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/genetics , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding/physiology , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species/metabolism , bcl-X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cell Death Differ ; 21(12): 1852-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25034783

ABSTRACT

The p62/SQSTM1 adapter protein has an important role in the regulation of several key signaling pathways and helps transport ubiquitinated proteins to the autophagosomes and proteasome for degradation. Here, we investigate the regulation and roles of p62/SQSTM1 during acute myeloid leukemia (AML) cell maturation into granulocytes. Levels of p62/SQSTM1 mRNA and protein were both significantly increased during all-trans retinoic acid (ATRA)-induced differentiation of AML cells through a mechanism that depends on NF-κB activation. We show that this response constitutes a survival mechanism that prolongs the life span of mature AML cells and mitigates the effects of accumulation of aggregated proteins that occurs during granulocytic differentiation. Interestingly, ATRA-induced p62/SQSTM1 upregulation was impaired in maturation-resistant AML cells but was reactivated when differentiation was restored in these cells. Primary blast cells of AML patients and CD34(+) progenitors exhibited significantly lower p62/SQSTM1 mRNA levels than did mature granulocytes from healthy donors. Our results demonstrate that p62/SQSTM1 expression is upregulated in mature compared with immature myeloid cells and reveal a pro-survival function of the NF-κB/SQSTM1 signaling axis during granulocytic differentiation of AML cells. These findings may help our understanding of neutrophil/granulocyte development and will guide the development of novel therapeutic strategies for refractory and relapsed AML patients with previous exposure to ATRA.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic/metabolism , Leukemia, Myeloid, Acute/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic/genetics , Gene Expression , Gene Expression Regulation, Leukemic , Granulocytes/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Sequestosome-1 Protein , Tretinoin/pharmacology , Ubiquitination , Up-Regulation
6.
Cell Death Dis ; 5: e1315, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24991767

ABSTRACT

Members of the WD-repeat protein interacting with phosphoinositides (WIPI) family are phosphatidylinositol 3-phosphate (PI3P) effectors that are essential for the formation of autophagosomes. Autophagosomes, unique double-membraned organelles, are characteristic for autophagy, a bulk degradation mechanism with cytoprotective and homeostatic function. Both, WIPI-1 and WIPI-2 are aberrantly expressed in several solid tumors, linking these genes to carcinogenesis. We now found that the expression of WIPI-1 was significantly reduced in a large cohort of 98 primary acute myeloid leukemia (AML) patient samples (complex karyotypes; t(8;21); t(15,17); inv(16)). In contrast, the expression of WIPI-2 was only reduced in acute promyelocytic leukemia (APL), a distinct subtype of AML (t(15,17)). As AML cells are blocked in their differentiation, we tested if the expression levels of WIPI-1 and WIPI-2 increase during all-trans retinoic acid (ATRA)-induced neutrophil differentiation of APL. According to the higher WIPI-1 expression in granulocytes compared with immature blast cells, WIPI-1 but not WIPI-2 expression was significantly induced during neutrophil differentiation of NB4 APL cells. Interestingly, the induction of WIPI-1 expression was dependent on the transcription factor PU.1, a master regulator of myelopoiesis, supporting our notion that WIPI-1 expression is reduced in AML patients lacking proper PU-1 activity. Further, knocking down WIPI-1 in NB4 cells markedly attenuated the autophagic flux and significantly reduced neutrophil differentiation. This result was also achieved by knocking down WIPI-2, suggesting that both WIPI-1 and WIPI-2 are functionally required and not redundant in mediating the PI3P signal at the onset of autophagy in NB4 cells. In line with these data, downregulation of PI3KC3 (hVPS34), which generates PI3P upstream of WIPIs, also inhibited neutrophil differentiation. In conclusion, we demonstrate that both WIPI-1 and WIPI-2 are required for the PI3P-dependent autophagic activity during neutrophil differentiation, and that PU.1-dependent WIPI-1 expression is significantly repressed in primary AML patient samples and that the induction of autophagic flux is associated with neutrophil differentiation of APL cells.


Subject(s)
Autophagy , Carrier Proteins/metabolism , Cell Differentiation , Leukemia, Promyelocytic, Acute/metabolism , Membrane Proteins/metabolism , Neutrophils/cytology , Adolescent , Adult , Aged , Autophagy-Related Proteins , Carrier Proteins/genetics , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Cohort Studies , Female , Gene Expression Regulation, Leukemic , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/physiopathology , Male , Membrane Proteins/genetics , Middle Aged , Neutrophils/metabolism , Phosphate-Binding Proteins , Young Adult
7.
Cell Death Dis ; 5: e1279, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24901050

ABSTRACT

Diffusely infiltrating gliomas are among the most prognostically discouraging neoplasia in human. Temozolomide (TMZ) in combination with radiotherapy is currently used for the treatment of glioblastoma (GBM) patients, but less than half of the patients respond to therapy and chemoresistance develops rapidly. Epigenetic silencing of the O(6)-methylguanine-DNA methyltransferase (MGMT) has been associated with longer survival in GBM patients treated with TMZ, but nuclear factor κB (NF-κB)-mediated survival signaling and TP53 mutations contribute significantly to TMZ resistance. Enhanced NF-κB is in part owing to downregulation of negative regulators of NF-κB activity, including Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) and NF-κB inhibitor interacting RAS-like 2 (NKIRAS2). Here we provide a novel mechanism independent of TP53 and MGMT by which oncogenic miR-125b confers TMZ resistance by targeting TNFAIP3 and NKIRAS2. GBM cells overexpressing miR-125b showed increased NF-κB activity and upregulation of anti-apoptotic and cell cycle genes. This was significantly associated with resistance of GBM cells to TNFα- and TNF-related inducing ligand-induced apoptosis as well as resistance to TMZ. Conversely, overexpression of anti-miR-125b resulted in cell cycle arrest, increased apoptosis and increased sensitivity to TMZ, indicating that endogenous miR-125b is sufficient to control these processes. GBM cells overexpressing TNFAIP3 and NKIRAS2 were refractory to miR-125b-induced apoptosis resistance as well as TMZ resistance, indicating that both genes are relevant targets of miR-125b. In GBM tissues, high miR-125b expression was significantly correlated with nuclear NF-κB confirming that miR-125b is implicated in NF-κB signaling. Most remarkably, miR-125b overexpression was clearly associated with shorter overall survival of patients treated with TMZ, suggesting that this microRNA is an important predictor of response to therapy.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Nuclear Proteins/metabolism , RNA, Neoplasm/pharmacology , ras Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Dacarbazine/pharmacology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , Temozolomide , Tumor Necrosis Factor alpha-Induced Protein 3 , ras Proteins/genetics
8.
Cell Death Differ ; 20(10): 1415-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23912709

ABSTRACT

p73, a member of the p53 tumor suppressor family, is involved in neurogenesis, sensory pathways, immunity, inflammation, and tumorigenesis. How p73 is able to participate in such a broad spectrum of different biological processes is still largely unknown. Here, we report a novel role of p73 in regulating lipid metabolism by direct transactivation of the promoter of autophagy-related protein 5 (ATG5), a gene whose product is required for autophagosome formation. Following nutrient deprivation, the livers of p73-deficient mice demonstrate a massive accumulation of lipid droplets, together with a low level of autophagy, suggesting that triglyceride hydrolysis into fatty acids is blocked owing to deficient autophagy (macrolipophagy). Compared with wild-type mice, mice functionally deficient in all the p73 isoforms exhibit decreased ATG5 expression and lower levels of autophagy in multiple organs. We further show that the TAp73α is the critical p73 isoform responsible for inducing ATG5 expression in a p53-independent manner and demonstrate that ATG5 gene transfer can correct autophagy and macrolipophagy defects in p73-deficient hepatocytes. These data strongly suggest that the p73-ATG5 axis represents a novel, key pathway for regulating lipid metabolism through autophagy. The identification of p73 as a major regulator of autophagy suggests that it may have an important role in preventing or delaying disease and aging by maintaining a homeostatic control.


Subject(s)
DNA-Binding Proteins/metabolism , Liver/physiology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Autophagy-Related Protein 5 , Cell Line, Tumor , DNA-Binding Proteins/genetics , HCT116 Cells , Humans , Lipid Metabolism/genetics , Liver/cytology , Liver/metabolism , Mice , Nuclear Proteins/genetics , Transcriptional Activation , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
9.
Oncogene ; 31(38): 4221-32, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22249264

ABSTRACT

The c-Src kinase regulates cancer cell invasion through inhibitor of DNA binding/differentiation 1 (ID1). Src and ID1 are frequently overexpressed in human lung adenocarcinoma. The current study aimed at identifying microRNAs (miRNAs) involved in the Src-ID1 signaling in lung cancer. Incubation of lung cancer cells with the Src inhibitor saracatinib led to the upregulation of several miRNAs including miR-29b, which was the most highly upregulated miRNA with predicted binding to the ID1 3'-untranslated region (UTR). Luciferase reporter assays confirmed direct binding of miR-29b to the ID1 3'-UTR. Expression of miR-29b suppressed ID1 levels and significantly reduced migration and invasion. Expression of antisense-miR-29b (anti-miR-29b), on the other hand, enhanced ID1 mRNA and protein levels, and significantly increased lung cancer cell migration and invasion, a hallmark of the Src-ID1 pathway. The ectopic expression of ID1 in miR-29b-overexpressing cells was able to rescue the migratory potential of these cells. Both, anti-miR-29b and ID1 overexpression diminished the effects of the Src inhibitors saracatinib and dasatinib on migration and invasion. Saracatinib and dasatinib decreased c-Myc transcriptional repression on miR-29b and led to increased ID1 protein levels, whereas forced expression of c-Myc repressed miR-29b and induced ID1. In agreement, we showed direct recruitment of c-Myc to the miR-29b promoter. miR-29b was significantly downregulated in primary lung adenocarcinoma samples compared with matched alveolar lung tissue, and miR-29b expression was a significant prognostic factor for patient outcome. These results suggest that miR-29b is involved in the Src-ID1 signaling pathway, is dysregulated in lung adenocarcinoma and is a potential predictive marker for Src kinase inhibitors.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Inhibitor of Differentiation Protein 1/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Signal Transduction , src-Family Kinases/genetics , 3' Untranslated Regions , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma of Lung , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Profiling , Genes, myc , Humans , Inhibitor of Differentiation Protein 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , MicroRNAs/metabolism , Prognosis , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
10.
J Intern Med ; 268(5): 410-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20964733

ABSTRACT

Autophagy (literally self-eating) is a catabolic mechanism involved in the recycling and turnover of cytoplasmic constituents. Although often referred to as type II programmed cell death, autophagy is primarily a survival rather than a cell death mechanism in response to different stress stimuli. Autophagy is a process in which part of the cytoplasm or entire organelles are sequestered into double-membrane vesicles, called autophagosomes, which ultimately fuse with lysosomes to degrade their contents. Studies show that autophagy is associated with a number of pathological conditions, including cancer, infectious diseases, myopathies and neurodegenerative disorders. With respect to cancer, it has been suggested that the early stages of tumourigenesis are associated with downregulation of autophagy-related (ATG) genes. Indeed, several ATG genes display tumour suppressor function, including Beclin1, which is frequently hemizygously deleted in breast cancer cells. Conversely, in advanced stages of tumourigenesis or during anticancer therapy, autophagy may promote survival of tumour cells in adverse environmental conditions. Therefore, a thorough understanding of autophagy in different cancer types and stages is a prerequisite to determine an autophagy-activating or autophagy-inhibiting treatment strategy.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/physiology , Neoplasms/therapy , Tumor Suppressor Protein p53/physiology , Humans , Models, Biological , Neoplasms/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism
12.
Oncogene ; 27(43): 5706-16, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18521079

ABSTRACT

Death-associated protein kinase 2 (DAPK2) belongs to a family of proapoptotic Ca(2+)/calmodulin-regulated serine/threonine kinases. We recently identified DAPK2 as an enhancing factor during granulocytic differentiation. To identify transcriptional DAPK2 regulators, we cloned 2.7 kb of the 5'-flanking region of the DAPK2 gene. We found that E2F1 and Krüppel-like factor 6 (KLF6) strongly activate the DAPK2 promoter. We mapped the E2F1 and KLF6 responsive elements to a GC-rich region 5' of exon 1 containing several binding sites for KLF6 and Sp1 but not for E2F. Moreover, we showed that transcriptional activation of DAPK2 by E2F1 and KLF6 is dependent on Sp1 using Sp1/KLF6-deficient insect cells, mithramycin A treatment to block Sp1-binding or Sp1 knockdown cells. Chromatin immunoprecipitation revealed recruitment of Sp1 and to lesser extent that of E2F1 and KLF6 to the DAPK2 promoter. Activation of E2F1 in osteosarcoma cells led to an increase of endogenous DAPK2 paralleled by cell death. Inhibition of DAPK2 expression resulted in significantly reduced cell death upon E2F1 activation. Similarly, KLF6 expression in H1299 cells increased DAPK2 levels accompanied by cell death that is markedly decreased upon DAPK2 knockdown. Moreover, E2F1 and KLF6 show cooperation in activating the DAPK2 promoter. In summary, our findings establish DAPK2 as a novel Sp1-dependent target gene for E2F1 and KLF6 in cell death response.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/genetics , E2F1 Transcription Factor/physiology , Kruppel-Like Transcription Factors/physiology , Proto-Oncogene Proteins/physiology , Apoptosis Regulatory Proteins/physiology , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cells, Cultured , Death-Associated Protein Kinases , Humans , Kruppel-Like Factor 6 , Promoter Regions, Genetic , Sp1 Transcription Factor/physiology
13.
Oncogene ; 27(24): 3489-93, 2008 May 29.
Article in English | MEDLINE | ID: mdl-18193090

ABSTRACT

The transcription factor PU.1 is essential for terminal myeloid differentiation, B- and T-cell development, erythropoiesis and hematopoietic stem cell maintenance. PU.1 functions as oncogene in Friend virus-induced erythroleukemia and as tumor suppressor in acute myeloid leukemias. Moreover, Friend virus-induced erythroleukemia requires maintenance of PU.1 expression and the disruption of p53 function greatly accelerates disease progression. It has been hypothesized that p53-mediated expression of the p21(Cip1) cell cycle inhibitor during differentiation of pre-erythroleukemia cells promotes selection against p53 function. In addition to the blockage of erythroblast differentiation provided by increased levels of PU.1, we propose that PU.1 alters p53 function. We demonstrate that PU.1 reduces the transcriptional activity of the p53 tumor suppressor family and thus inhibits activation of genes important for cell cycle regulation and apoptosis. Inhibition is mediated through binding of PU.1 to the DNA-binding and/or oligomerization domains of p53/p73 proteins. Lastly, knocking down endogenous PU.1 in p53 wild-type REH B-cell precursor leukemia cells leads to increased expression of the p53 target p21(Cip1).


Subject(s)
Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunoprecipitation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Isoforms , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcriptional Activation , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Gene Ther ; 13(20): 1480-92, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16738691

ABSTRACT

CCR5 is the chemokine co-receptor for R5-tropic human immunodeficiency virus type 1 (HIV-1) isolates most often associated with primary infection. We have developed an HIV-1 self-inactivating vector, CAD-R5, containing a CCR5 single-chain antibody (intrabody) gene, which when expressed in T-cell lines and primary CD4+ T cells disrupts CCR5 cell surface expression and provides protection from R5-tropic isolate exposure. Furthermore, CAD-R5 intrabody expression in primary CD4+ T cells supports significant growth and enrichment over time during HIV-1-pulsed dendritic cell-T-cell interactions. These results indicate that CCR5 intrabody-expressing CD4+ T cells are refractory against this highly efficient primary route of infection. CD34+ cells transduced with the CAD-R5 vector gave rise to CD4+ and CD8+ thymocytes in non-obese diabetic (NOD)/ severely combined-immunodeficient (SCID)-human thymus/liver (hu thy/liv) mice, suggesting that CCR5 intrabody expression can be maintained throughout differentiation without obvious cellular effects. CD4+ T cells isolated from NOD/SCID-hu thy/liv mice were resistant to R5-tropic HIV-1 challenge demonstrating the maintenance of protection. Our findings demonstrate delivery of anti-HIV-1 activity through CCR5 intrabodies in primary CD4+ T cells and CD34+ cell-derived T-cell progeny. Thus, gene delivery strategies that provide a selective survival and growth advantage for T effector cells may provide a therapeutic benefit for HIV-1-infected individuals who have failed conventional therapies.


Subject(s)
Antibodies/genetics , CD4-Positive T-Lymphocytes/immunology , Genetic Therapy/methods , HIV Infections/therapy , HIV-1/physiology , Receptors, CCR5/genetics , Animals , Antibodies/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Cytoprotection , Dendritic Cells/immunology , Dendritic Cells/virology , Gene Expression Regulation , HIV Infections/immunology , Humans , Mice , Mice, SCID , Receptors, CCR5/metabolism
16.
Oncogene ; 25(14): 2030-9, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16301995

ABSTRACT

The tumor suppressor gene hypermethylated in cancer 1 (HIC1), located on human chromosome 17p13.3, is frequently silenced in cancer by epigenetic mechanisms. Hypermethylated in cancer 1 belongs to the bric à brac/poxviruses and zinc-finger family of transcription factors and acts by repressing target gene expression. It has been shown that enforced p53 expression leads to increased HIC1 mRNA, and recent data suggest that p53 and Hic1 cooperate in tumorigenesis. In order to elucidate the regulation of HIC1 expression, we have analysed the HIC1 promoter region for p53-dependent induction of gene expression. Using progressively truncated luciferase reporter gene constructs, we have identified a p53-responsive element (PRE) 500 bp upstream of the TATA-box containing promoter P0 of HIC1, which is sequence specifically bound by p53 in vitro as assessed by electrophoretic mobility shift assays. We demonstrate that this HIC1 p53-responsive element (HIC1.PRE) is necessary and sufficient to mediate induction of transcription by p53. This result is supported by the observation that abolishing endogenous wild-type p53 function prevents HIC1 mRNA induction in response to UV-induced DNA damage. Other members of the p53 family, notably TAp73beta and DeltaNp63alpha, can also act through this HIC1.PRE to induce transcription of HIC1, and finally, hypermethylation of the HIC1 promoter attenuates inducibility by p53.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , DNA Primers , Electrophoretic Mobility Shift Assay , Genes, Reporter , Genes, p53 , Humans , Kruppel-Like Transcription Factors , Molecular Sequence Data , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Up-Regulation
17.
Leuk Lymphoma ; 42(5): 1077-87, 2001.
Article in English | MEDLINE | ID: mdl-11697625

ABSTRACT

The p16INK4a gene is often disrupted or transcriptionally silenced by CpG island methylation in human cancers. However, in acute myeloid leukaemia (AML) alterations of the INK4a-ARF tumour suppressor locus are rarely found despite the noted variable p16INK4a mRNA and protein levels. The p14ARF, an alternative reading frame protein encoded from the same INK4a-ARF locus, is a potent tumour suppressor functionally linked to p53. There is little known regarding the role of p14ARF in primary human tumours. Therefore, we analysed the expression patterns of these two tumour suppressors in 37 cases of AML. The relative expression of p16INK4a and p14ARF mRNA in AML blasts, measured by a specific p16INK4a/p14ARF multiplex RT-PCR, was significantly shifted towards p14ARF whereas relatively lower levels of p16INK4a were detected. Quantitative RT-PCR revealed significantly higher expression of both transcripts in AML blasts when compared to normal differentiated myeloid cells or CD34+ progenitor cells. Furthermore, a good correlation between p16INK4a protein and mRNA was observed, whereas no correlation was found with p14ARF. Our results suggest: a) increased levels of both p16INK4a and p14ARF may participate in the pathogenesis of AML, b) that high p14ARF mRNA expression might influence p16INK4a transcription and c) that post-transcriptional regulatory mechanisms are important for p14ARF expression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Leukemia, Myeloid/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Acute Disease , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/pathology , Leukocytes/cytology , Leukocytes/metabolism , RNA, Neoplasm/analysis , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/metabolism , Tumor Suppressor Protein p14ARF/genetics
18.
Ann Oncol ; 12(7): 981-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11521806

ABSTRACT

The p73 protein shares structural and functional similarities with the tumour-suppressor p53, but its role in neoplastic transformation is unknown. Alternative splicing leads to the expression of at least nine p73 C-terminal mRNA splice variants (alpha, beta, gamma, delta, epsilon, zeta, eta, eta1, theta). In this survey, we analyse the expression of p73 by real-time quantitative RT-PCR, its known C-terminal variants with an RT-PCR-Southern technique and by Western blot in samples of 51 patients with B-CLL, normal B lymphocytes from eight individuals, and five haematopoetic cell lines. p73alpha protein expression positively correlated with higher risk B-CLL stages (P = 0.046). Total p73 mRNA expression was higher (P = 0.01) and p73alpha protein more frequently detected (P = 0.008) in B-CLL compared with normal CD19+-B-lymphocytes. p73 C-terminal mRNA variants were expressed both in B-CLL and in normal B-lymphocytes, but their expression was biased since the gamma (P = 0.041), the theta (P < 0.001), and the eta variant (P = 0.033) prevailed in normal B-lymphocytes. In summary, we conclude that the accumulation of p73, the expression pattern of particular p73 variants and its link to progression may play a distinct role in the molecular pathology B-CLL.


Subject(s)
Biomarkers, Tumor/analysis , DNA-Binding Proteins/analysis , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Nuclear Proteins/analysis , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Male , Middle Aged , Nuclear Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Tumor Protein p73 , Tumor Suppressor Proteins , Up-Regulation
19.
Biochem Biophys Res Commun ; 277(1): 62-5, 2000 Oct 14.
Article in English | MEDLINE | ID: mdl-11027640

ABSTRACT

The p53 homologue p73 is expressed in at least six different isoforms (alpha, beta, gamma, delta, epsilon, and zeta), but unlike p53 it has rarely been found mutated in human cancers. However, altered expression of this gene has been reported in cancer cells. In order to understand if p73 is involved in normal and malignant development of myeloid cells, we investigated the expression pattern of the different p73 isoforms in progenitor and mature normal myeloid cells as well as in cells derived from acute and chronic myeloid leukemias. The results show that expression of p73 is markedly enhanced during differentiation of myeloid leukemic cells and that leukemic blasts from patients show an increased expression of the shorter p73 isoforms (gamma, delta, epsilon, zeta). In particular the epsilon isoform is only expressed in leukemic cells and completely absent in mature myeloid cells. Altogether our data suggest that p73 is involved in myeloid differentiation and its altered expression is involved in leukemic degeneration.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Nuclear Proteins/genetics , Acute Disease , Blotting, Southern , Blotting, Western , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor , HL-60 Cells , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid/genetics , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
20.
Int J Cancer ; 88(1): 66-70, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-10962441

ABSTRACT

The p73 gene encodes a protein with substantial structural and functional similarities to the tumour-suppressor p53. Alternative splicing of p73 mRNA leads to expression of 6 known RNA species and proteins (alpha, beta, gamma, delta, epsilon, zeta). We analysed the expression of these splice variants in ovarian adenocarcinoma by RT-PCR followed by detection of amplicons with the Southern technique and by immunoblot in 32 malignant and benign epithelial ovarian tumour specimens and 3 ovarian adenocarcinoma cell lines (A2780, 2008, OVCAR-3). p73alpha mRNA was expressed in all 17 ovarian cancer specimens, and 14 of 17 expressed at least 3 splice variants. In contrast, a different expression pattern was present in the ovarian adenomas: p73alpha was detected in 6 of 12 benign tumours, and only 1 adenoma expressed 3 splice variants. p73 protein was expressed in 9 of 16 ovarian cancer specimens, in all cell lines and in 1 of 3 borderline tumours. In contrast, none of 9 ovarian adenomas expressed detectable amounts of p73 protein. Expression of p73 mRNA and protein was not correlated with FIGO stage and histological grade, but we observed a significant correlation with over-expression of p53 protein. In summary, epithelial ovarian cancers express a more complex p73 isoform pattern and higher levels of p73 mRNA and protein than ovarian adenomas.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Alternative Splicing , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Ovarian Neoplasms/metabolism , Adenocarcinoma/genetics , Adenoma/genetics , Adult , Aged , Aged, 80 and over , Female , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/genetics , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL