Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Pineal Res ; 70(3): e12724, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33615553

ABSTRACT

Cancer-related fatigue (CRF) and stress are common symptoms in cancer patients and represent early side effects of cancer treatment which affect the life quality of the patients. CRF may partly depend on disruption of the circadian rhythm. Locomotor activity and corticosterone rhythms are two important circadian outputs which can be used to analyze possible effects on the circadian function during cancer development and treatment. The present study analyzes the relationship between locomotor activity rhythm, corticosterone levels, hepatocellular carcinoma (HCC) development, and radiotherapy treatment in a mouse model. HCC was induced in mice by single injection of diethylnitrosamine (DEN) and chronic treatment of phenobarbital in drinking water. Another group received chronic phenobarbital treatment only. Tumor bearing animals were divided randomly into four groups irradiated at four different Zeitgeber time points. Spontaneous locomotor activity was recorded continuously; serum corticosterone levels and p-ERK immunoreaction in the suprachiasmatic nucleus (SCN) were investigated. Phenobarbital treated mice showed damped corticosterone levels and a less stable 24 hours activity rhythm as well as an increase in activity during the light phase, reminiscent of sleep disruption. The tumor mice showed an increase in corticosterone level during the inactive phase and decreased activity during the dark phase, reminiscent of CRF. After irradiation, corticosterone levels were further increased and locomotor activity rhythms were disrupted. Lowest corticosterone levels were observed after irradiation during the early light phase; thus, this time might be the best to apply radiotherapy in order to minimize side effects.


Subject(s)
Activity Cycles , Behavior, Animal , Carcinoma, Hepatocellular/radiotherapy , Circadian Rhythm , Corticosterone/blood , Liver Neoplasms, Experimental/radiotherapy , Locomotion , Suprachiasmatic Nucleus/physiopathology , Animals , Biomarkers/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/physiopathology , Chronotherapy , Diethylnitrosamine , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms, Experimental/blood , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Period Circadian Proteins/genetics , Phenobarbital , Phosphorylation , Suprachiasmatic Nucleus/metabolism , Time Factors
2.
Haematologica ; 104(1): 35-46, 2019 01.
Article in English | MEDLINE | ID: mdl-30093397

ABSTRACT

The homeobox gene HLXB9 encodes for the transcription factor HB9, which is essential for pancreatic as well as motor neuronal development. Beside its physiological expression pattern, aberrant HB9 expression has been observed in several neoplasias. Especially in infant translocation t(7;12) acute myeloid leukemia, aberrant HB9 expression is the only known molecular hallmark and is assumed to be a key factor in leukemic transformation. However, so far, only poor functional data exist addressing the oncogenic potential of HB9 or its influence on hematopoiesis. We investigated the influence of HB9 on cell proliferation and cell cycle in vitro, as well as on hematopoietic stem cell differentiation in vivo using murine and human model systems. In vitro, HB9 expression led to premature senescence in human HT1080 and murine NIH3T3 cells, providing for the first time evidence for an oncogenic potential of HB9. Onset of senescence was characterized by induction of the p53-p21 tumor suppressor network, resulting in growth arrest, accompanied by morphological transformation and expression of senescence-associated ß-galactosidase. In vivo, HB9-transduced primary murine hematopoietic stem and progenitor cells underwent a profound differentiation arrest and accumulated at the megakaryocyte/erythrocyte progenitor stage. In line, gene expression analyses revealed de novo expression of erythropoiesis-related genes in human CD34+hematopoietic stem and progenitor cells upon HB9 expression. In summary, the novel findings of HB9-dependent premature senescence and myeloid-biased perturbed hematopoietic differentiation, for the first time shed light on the oncogenic properties of HB9 in translocation t(7;12) acute myeloid leukemia.


Subject(s)
Cell Cycle , Cell Differentiation , Cellular Senescence , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/biosynthesis , Transcription Factors/biosynthesis , Animals , Erythropoiesis/genetics , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , NIH 3T3 Cells , Neoplasm Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic
3.
J Biol Chem ; 292(20): 8331-8341, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28348086

ABSTRACT

The cyclin-dependent kinase inhibitor p21 is an important player in stress pathways exhibiting both tumor-suppressive and oncogenic functions. Thus, expression of p21 has to be tightly controlled, which is achieved by numerous mechanisms at the transcriptional, translational, and posttranslational level. Performing immunoprecipitation of bromouridine-labeled p21 mRNAs that had been incubated before with cytoplasmic extracts of untreated HCT116 colon carcinoma cells, we identified the DEAD-box RNA helicase DDX41 as a novel regulator of p21 expression. DDX41 specifically precipitates with the 3'UTR, but not with the 5'UTR, of p21 mRNA. Knockdown of DDX41 increases basal and γ irradiation-induced p21 protein levels without affecting p21 mRNA expression. Conversely, overexpression of DDX41 strongly inhibits expression of a FLAG-p21 and a luciferase construct, but only in the presence of the p21 3'UTR. Together, these data suggest that this helicase regulates p21 expression at the translational level independent of the transcriptional activity of p53. However, knockdown of DDX41 completely fails to increase p21 protein levels in p53-deficient HCT116 cells. Moreover, posttranslational up-regulation of p21 achieved in both p53+/+ and p53-/- HCT116 cells in response to pharmaceutical inhibition of the proteasome (by MG-132) or p90 ribosomal S6 kinases (by BI-D1870) is further increased by knockdown of DDX41 only in p53-proficient but not in p53-deficient cells. Although our data demonstrate that DDX41 suppresses p21 translation without disturbing the function of p53 to directly induce p21 mRNA expression, this process indirectly requires p53, perhaps in the form of another p53 target gene or as a still undefined posttranscriptional function of p53.


Subject(s)
3' Untranslated Regions/physiology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DEAD-box RNA Helicases/metabolism , Protein Biosynthesis/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DEAD-box RNA Helicases/genetics , Gene Knockdown Techniques , Humans , Protein Biosynthesis/drug effects , Pteridines/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Support Care Cancer ; 24(10): 4241-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27165055

ABSTRACT

BACKGROUND: The mechanisms of skin aging have not been completely elucidated. Anecdotal data suggests that EGFR inhibition accelerates aging-like skin changes. OBJECTIVE: The objective of the study was to evaluate the clinical characteristics and investigate the cellular and molecular mechanisms underlying skin changes associated with the use of EFGRIs. PATIENTS AND METHODS: Patients during prolonged treatment with EGFRIs (>3 months) were analyzed for aging-like skin changes. Baseline EGFR expression was compared in young (<25 years old) vs. old (> 65 years old) skin. In addition, the regulation of extracellular matrix, senescence-associated genes, and cell cycle status was measured in primary human keratinocytes treated with erlotinib in vitro. RESULTS: There were progressive signs of skin aging, including xerosis cutis, atrophy, rhytide formation, and/or actinic purpura in 12 patients. Keratinocytes treated with erlotinib in vitro showed a significant down-modulation of hyaluronan synthases (HAS2 and HAS3), whereas senescence-associated genes (p21, p53, IL-6, maspin) were upregulated, along with a G1 cell cycle arrest and stronger SA ß-Gal activity. There was significantly decreased baseline expression in EGFR density in aged skin, when compared to young controls. CONCLUSIONS: EGFR inhibition results in molecular alterations in keratinocytes that may contribute to the observed skin aging of patients treated with respective targeted agents.


Subject(s)
ErbB Receptors/genetics , Erlotinib Hydrochloride/therapeutic use , Skin Aging/genetics , Skin Diseases/genetics , Aged , Aged, 80 and over , Aging , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/pharmacology , Female , Humans , Retrospective Studies
5.
Cell Death Dis ; 15(5): 373, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811535

ABSTRACT

The targeted elimination of radio- or chemotherapy-induced senescent cells by so-called senolytic substances represents a promising approach to reduce tumor relapse as well as therapeutic side effects such as fibrosis. We screened an in-house library of 178 substances derived from marine sponges, endophytic fungi, and higher plants, and determined their senolytic activities towards DNA damage-induced senescent HCT116 colon carcinoma cells. The Pan-PI3K-inhibitor wortmannin and its clinical derivative, PX-866, were identified to act as senolytics. PX-866 potently induced apoptotic cell death in senescent HCT116, MCF-7 mammary carcinoma, and A549 lung carcinoma cells, independently of whether senescence was induced by ionizing radiation or by chemotherapeutics, but not in proliferating cells. Other Pan-PI3K inhibitors, such as the FDA-approved drug BAY80-6946 (Copanlisib, Aliqopa®), also efficiently and specifically eliminated senescent cells. Interestingly, only the simultaneous inhibition of both PI3K class I alpha (with BYL-719 (Alpelisib, Piqray®)) and delta (with CAL-101 (Idelalisib, Zydelig®)) isoforms was sufficient to induce senolysis, whereas single application of these inhibitors had no effect. On the molecular level, inhibition of PI3Ks resulted in an increased proteasomal degradation of the CDK inhibitor p21WAF1/CIP1 in all tumor cell lines analyzed. This led to a timely induction of apoptosis in senescent tumor cells. Taken together, the senolytic properties of PI3K-inhibitors reveal a novel dimension of these promising compounds, which holds particular potential when employed alongside DNA damaging agents in combination tumor therapies.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21 , Humans , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , HCT116 Cells , Proteasome Endopeptidase Complex/metabolism , Apoptosis/drug effects , Phosphoinositide-3 Kinase Inhibitors/pharmacology , MCF-7 Cells , Proteolysis/drug effects , A549 Cells , Wortmannin/pharmacology , Senotherapeutics/pharmacology , Class I Phosphatidylinositol 3-Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , DNA Damage/drug effects , Pyrimidines , Quinazolines
6.
Int J Cancer ; 133(11): 2551-62, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23686572

ABSTRACT

The p14(ARF) tumor suppressor triggers cell death or cell cycle arrest upon oncogenic stress. In MCF-7 breast carcinoma cells, expression of the tumor suppressor gene p14(ARF) fails to trigger apoptosis but induces an arrest in the G1 and, to a lesser extent, in the G2 phase in the cell division cycle. Here, inhibition of cell cycle arrest resulted in apoptosis induction in caspase-3 proficient MCF-7 cells upon expression of p14(ARF) . This occurred in the absence of S-phase progression or mitotic entry. In contrast, syngeneic, caspase-3-deficient MCF-7 cells remained entirely resistant to p14(ARF) -induced apoptosis. Thus, cell cycle checkpoint abrogation overcomes resistance to p14(ARF) -induced cell death and promotes cell death via a caspase-3-dependent pathway. Cell death coincided with dissipation of the mitochondrial membrane potential, release of cytochrome c, and was inhibitable by pan-caspase inhibitors and the caspase-3/7 inhibitor zDEVD-fmk. Of note, mitochondrial events of apoptosis execution depended entirely on caspase-3 proficiency indicating that caspase-3 either acts "up-stream" of the mitochondria in a "non-canonical" pathway or mediates a mitochondrial feedback loop to amplify the apoptotic caspase signal in p14(ARF) -induced stress signaling.


Subject(s)
Apoptosis/genetics , Caspase 3/metabolism , Mitochondria/metabolism , Tumor Suppressor Protein p14ARF/genetics , Breast Neoplasms , Cell Cycle Checkpoints/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mitochondria/genetics , Signal Transduction , Tumor Suppressor Protein p14ARF/metabolism
7.
Cancer Med ; 10(21): 7712-7725, 2021 11.
Article in English | MEDLINE | ID: mdl-34545699

ABSTRACT

This study investigates whether a chronotherapeutic treatment of hepatocellular carcinoma (HCC) may improve treatment efficacy and mitigate side effects on non-tumoral liver (NTL). HCC was induced in Per2::luc mice which were irradiated at four time points of the day. Proliferation and DNA-double strand breaks were analyzed in irradiated and nonirradiated animals by detection of Ki67 and γ-H2AX. Prior to whole animal experiments, organotypic slice cultures were investigated to determine the dosage to be used in whole animal experiments. Irradiation was most effective at the proliferation peaks in HCC at ZT02 (early inactivity phase) and ZT20 (late activity phase). Irradiation effects on NTL were minimal at ZT20. As compared with NTL, nonirradiated HCC revealed disruption in daily variation and downregulation of all investigated clock genes except Per1. Irradiation affected rhythmic clock gene expression in NTL and HCC at all ZTs except at ZT20 (late activity phase). Irradiation at ZT20 had no effect on total leukocyte numbers. Our results indicate ZT20 as the optimal time point for irradiation of HCC in mice at which the ratio between efficacy of tumor treatment and toxic side effects was maximal. Translational studies are now needed to evaluate whether the late activity phase is the optimal time point for irradiation of HCC in man.


Subject(s)
Carcinoma, Hepatocellular/radiotherapy , Chronotherapy , Liver Neoplasms/radiotherapy , Animals , Blood Cell Count , CLOCK Proteins/genetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , DNA Damage , Down-Regulation , Gene Expression , Histones/analysis , Ki-67 Antigen/analysis , Liver Neoplasms/blood , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Time Factors
8.
Cell Death Differ ; 27(4): 1274-1285, 2020 04.
Article in English | MEDLINE | ID: mdl-31511650

ABSTRACT

In recent years it has become more and more apparent that the regulation of gene expression by RNA-binding proteins (RBPs) is of utmost importance for most cellular signaling pathways. RBPs control several aspects of RNA biogenesis including splicing, localization, stability, and translation efficiency. One of these RBPs is RBM47 that recently has been suggested to function as a tumor suppressor as it was shown to suppress breast and colon cancer progression. Here we demonstrate that RBM47 is an important regulator of basal and DNA damage-induced p53 and p21WAF1/CIP1 protein expression. Knockdown of RBM47 by siRNAs results in a strong reduction in p53 mRNA and protein levels due to an impaired p53 promoter activity. Accordingly, overexpression of Flag-RBM47 enhances p53 promoter activity demonstrating that RBM47 regulates p53 at the transcriptional level. By controlling p53, knockdown of RBM47 concomitantly decreases also p21 expression at the transcriptional level, driving irradiated carcinoma cell lines from different entities into cell death rather than into senescence. Thus, RBM47 represents a novel molecular switch of cell fate decisions that functions as a regulator of the p53/p21-signaling axis.


Subject(s)
Cell Lineage/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , Tumor Suppressor Protein p53/metabolism
9.
Cell Cycle ; 19(4): 464-478, 2020 02.
Article in English | MEDLINE | ID: mdl-31959066

ABSTRACT

Microtubule-targeting agents (MTAs) are the most effective chemotherapeutics used in cancer therapy to date, but their clinical use is often hampered by the acquisition of resistance. Thereby, elucidation of the molecular signaling pathways activated by novel FDA-approved MTAs such as eribulin is important for future therapeutic applications. In contrast to several reports, we show here that regardless of the presence of caspase-3, clinically relevant concentrations of eribulin and the classical MTA paclitaxel predominantly induce caspase-independent cell death in MCF-7 breast carcinoma cells. On the molecular level, several key proteins involved in apoptosis such as p53, Plk1, caspase-2, and Bim as well as the two MAPKs ERK and JNK were activated by both compounds to a similar extent. However, none of them proved to be important for eribulin- and paclitaxel-induced cytotoxicity, as their siRNA-mediated knockdown or inactivation by small molecule inhibitors did not alter cell death rates. In contrast, knockdown of the anti-apoptotic Bcl-2 protein, which becomes heavily phosphorylated at Ser70 during MTA treatment, resulted surprisingly in a reduction of MTA-mediated cell death. This phenomenon can be most likely explained by our observation that the absence of Bcl-2 slowed down cell cycle progression resulting in fewer cells entering mitosis, thereby delaying the mitotic capability of these MTAs to induce cell death. Taken together, although eribulin and paclitaxel disturb the mitotic spindle differently, they exhibit no functional differences in downstream molecular cell death signaling in MCF-7 breast cancer cells.


Subject(s)
Caspase 3/metabolism , Furans/pharmacology , Ketones/pharmacology , Microtubules/metabolism , Paclitaxel/pharmacology , Signal Transduction , Anthracenes/pharmacology , Butadienes/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Humans , Microtubules/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects
10.
Cells ; 9(9)2020 08 24.
Article in English | MEDLINE | ID: mdl-32846968

ABSTRACT

The stress-inducible and senescence-associated tumor suppressor SIRT4, a member of the family of mitochondrial sirtuins (SIRT3, SIRT4, and SIRT5), regulates bioenergetics and metabolism via NAD+-dependent enzymatic activities. Next to the known mitochondrial location, we found that a fraction of endogenous or ectopically expressed SIRT4, but not SIRT3, is present in the cytosol and predominantly localizes to centrosomes. Confocal spinning disk microscopy revealed that SIRT4 is found during the cell cycle dynamically at centrosomes with an intensity peak in G2 and early mitosis. Moreover, SIRT4 precipitates with microtubules and interacts with structural (α,ß-tubulin, γ-tubulin, TUBGCP2, TUBGCP3) and regulatory (HDAC6) microtubule components as detected by co-immunoprecipitation and mass spectrometric analyses of the mitotic SIRT4 interactome. Overexpression of SIRT4 resulted in a pronounced decrease of acetylated α-tubulin (K40) associated with altered microtubule dynamics in mitotic cells. SIRT4 or the N-terminally truncated variant SIRT4(ΔN28), which is unable to translocate into mitochondria, delayed mitotic progression and reduced cell proliferation. This study extends the functional roles of SIRT4 beyond mitochondrial metabolism and provides the first evidence that SIRT4 acts as a novel centrosomal/microtubule-associated protein in the regulation of cell cycle progression. Thus, stress-induced SIRT4 may exert its role as tumor suppressor through mitochondrial as well as extramitochondrial functions, the latter associated with its localization at the mitotic spindle apparatus.


Subject(s)
Centrosome/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitosis/genetics , Sirtuins/metabolism , Humans
11.
Biol Chem ; 390(10): 951-63, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19453282

ABSTRACT

Upon DNA damage and other stresses, the transcription factor p53 elicits numerous responses including DNA repair, cell cycle arrest and apoptosis, properties that make p53 the prototype tumor suppressor. In addition, p53 also transactivates genes whose products act in an anti-apoptotic manner providing strong evidence that p53 exhibits both tumor suppressive and tumorigenic functions. Although several events were postulated to contribute to the p53-mediated decision process, the precise mechanism(s) that governs p53 activities is still elusive. Recently, it was found that the p53 gene allows expression of at least nine different isoforms that arise from multiple splicing events and the usage of alternative promoters. Several of these isoforms were shown to critically interfere with the function of the full-length p53 mainly by acting in a dominant-negative manner. However, an isoform-dependent selective activation of p53 target genes was also observed. Furthermore, certain p53 isoforms are aberrantly expressed in various tumors strongly implying their involvement in tumorigenic events. Thus, p53 isoforms may represent crucial determinants in p53-mediated decision processes whose precise functions (their do's and don'ts) are only beginning to emerge.


Subject(s)
Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Cycle/genetics , Cell Cycle/physiology , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , Humans , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Tumor Suppressor Protein p53/genetics
12.
Mol Cell Biol ; 26(5): 1967-78, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16479014

ABSTRACT

Due to their tremendous apoptosis-inducing potential, proteasomal inhibitors (PIs) have recently entered clinical trials. Here we show, however, that various PIs rescued proliferating tumor cells from death receptor-induced apoptosis. This protection correlated with the stabilization of X-linked IAP (XIAP) and c-FLIP and the inhibition of caspase activation. Together with the observation that PIs could not protect cells expressing XIAP or c-FLIP short interfering RNAs (siRNAs) from death receptor-induced apoptosis, our results demonstrate that PIs mediate their protective effect via the stabilization of these antiapoptotic proteins. Furthermore, we show that once these proteins were eliminated, either by long-term treatment with death receptor ligands or by siRNA-mediated suppression, active caspases accumulated to an even larger extent in the presence of PIs. Together, our data support a biphasic role for the proteasome in apoptosis, as they show that its constitutive activity is crucial for the rapid initiation of the death program by eliminating antiapoptotic proteins, whereas at later stages, the proteasome acts in an antiapoptotic manner due to the proteolysis of caspases. Thus, for a successful PI-based tumor therapy, it is crucial to carefully evaluate basal proteasomal activity and the status of antiapoptotic proteins, as their PI-mediated prolonged stability might even cause adverse effects, leading to the survival of a tumor.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Enzyme Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspases/drug effects , Caspases/metabolism , Death Domain Receptor Signaling Adaptor Proteins , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leupeptins/pharmacology , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Proteasome Inhibitors , RNA, Small Interfering , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/drug effects , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , fas Receptor/drug effects , fas Receptor/metabolism
13.
Mol Cell Biol ; 25(7): 2808-18, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15767684

ABSTRACT

Although signaling by death receptors involves the recruitment of common components into their death-inducing signaling complexes (DISCs), apoptosis susceptibility of various tumor cells to each individual receptor differs quite dramatically. Recently it was shown that, besides caspase-8, caspase-10 is also recruited to the DISCs, but its function in death receptor signaling remains unknown. Here we show that expression of caspase-10 sensitizes MCF-7 breast carcinoma cells to TRAIL- but not tumor necrosis factor (TNF)-induced apoptosis. This sensitization is most obvious at low TRAIL concentrations or when apoptosis is assessed at early time points. Caspase-10-mediated sensitization for TRAIL-induced apoptosis appears to be dependent on caspase-3, as expression of caspase-10 in MCF-7/casp-3 cells but not in caspase-3-deficient MCF-7 cells overcomes TRAIL resistance. Interestingly, neutralization of TRAIL receptor 2 (TRAIL-R2), but not TRAIL-R1, impaired apoptosis in a caspase-10-dependent manner, indicating that caspase-10 enhances TRAIL-R2-induced cell death. Furthermore, whereas processing of caspase-10 was delayed in TNF-treated cells, TRAIL triggered a very rapid activation of caspase-10 and -3. Therefore, we propose a model in which caspase-10 is a crucial component during TRAIL-mediated apoptosis that in addition actively requires caspase-3. This might be especially important in systems where only low TRAIL concentrations are supplied that are not sufficient for the fast recruitment of caspase-8 to the DISC.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Caspases/metabolism , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Breast Neoplasms/metabolism , Caspase 10 , Caspase 3 , Caspase 8 , Caspases/deficiency , Caspases/genetics , Cell Line, Tumor , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Kinetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factors/pharmacology
14.
Cancer Res ; 66(23): 11254-62, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145870

ABSTRACT

The role of the cyclin-dependent kinase (CDK) inhibitor p21 as a mediator of p53-induced growth arrest is well established. In addition, recent data provide strong evidence for new emerging functions of p21, including a role as a modulator of apoptosis. The mechanisms, however, by which p21 interferes with the death machinery, especially following ionizing radiation (IR), are largely unknown. Here, we report that IR induced caspase-9 and caspase-3 activation and subsequent apoptosis only in p21-deficient colon carcinoma cells, whereas similar treated wild-type cells were permanently arrested in the G(2)-M phase, correlating with the induction of cellular senescence. Interestingly, activation of the mitochondrial pathway, including caspase-2 processing, depolarization of the outer mitochondrial membrane, and cytochrome c release, was achieved by IR in both cell lines, indicating that p21 inhibits an event downstream of mitochondria but preceding caspase-9 activation. IR-induced p21 protein expression was restricted to the nucleus, and no evidence for a mitochondrial or cytoplasmic association was found. In addition, p21 did neither interact with caspase-3 or caspase-9, suggesting that these events are not required for the observed protection. Consistent with this assumption, we found that CDK inhibitors potently abrogated IR-induced caspase processing and activation without affecting mitochondrial events. In addition, in vitro caspase activation assays yielded higher caspase-3 activities in extracts of irradiated p21-deficient cells compared with extracts of similar treated wild-type cells. Thus, our results strongly indicate that p21 protects cells from IR-induced apoptosis by suppression of CDK activity that seems to be required for activation of the caspase cascade downstream of the mitochondria.


Subject(s)
Apoptosis/radiation effects , Caspase 9/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/metabolism , Mitochondria/radiation effects , Blotting, Western , Caspase 3/metabolism , Caspase Inhibitors , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Genotype , HCT116 Cells , Humans , Membrane Potentials/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Mutation/genetics , Oligopeptides/pharmacology , Protein Binding/radiation effects , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Radiation, Ionizing , Roscovitine , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Astrobiology ; 17(2): 136-144, 2017 02.
Article in English | MEDLINE | ID: mdl-28206821

ABSTRACT

This study addresses the viability of the lichen Xanthoria elegans after high-dose ionizing irradiation in the frame of the STARLIFE campaign. The first set of experiments was intended to resemble several types of galactic cosmic radiation (GCR) as present beyond the magnetic shield of Earth. In the second set of experiments, γ radiation up to 113 kGy was applied to test the limit of lichen resistance to ionizing radiation. Entire thalli of Xanthoria elegans were irradiated in the anhydrobiotic state. After STARLIFE 1, the metabolic activity of both symbionts was quantified by live/dead staining with confocal laser scanning microscopy. The photosynthetic activity was measured after the respective irradiation to assess the ability of the symbiotic green algae to restore photosynthesis after irradiation. The STARLIFE campaign complements the results of the LIFE experiments at the EXPOSE-E facility on the International Space Station by testing the model organism Xanthoria elegans on its resistance to hazardous radiation that might be accumulated during long-term space exposure. In addition, the photosynthetic activity of metabolically active lichen was investigated after X-ray irradiation up to 100 Gy (3.3 Gy/min). Since previous astrobiological experiments were mostly performed with anhydrobiotic lichen, these experiments will broaden our knowledge on the correlation of physiological state and astrobiological stressors. Key Words: Astrobiology-Extremotolerance-Gamma rays-Ionizing radiation-Lichens-Viability. Astrobiology 17, 136-144.


Subject(s)
Cosmic Radiation , Lichens/radiation effects , Radiation, Ionizing , Space Simulation , Dose-Response Relationship, Radiation , Helium/chemistry , Ions , Iron/chemistry , Lichens/metabolism , Microscopy, Confocal , Photosynthesis/radiation effects , X-Rays
16.
Brain Res ; 1100(1): 1-12, 2006 Jul 19.
Article in English | MEDLINE | ID: mdl-16781680

ABSTRACT

The choroid plexus epithelium constitutes the structural basis of the blood-cerebrospinal fluid barrier. We previously demonstrated that Streptococcus suis (S. suis), a relevant cause of bacterial meningitis in pigs and humans, affects porcine choroid plexus epithelial cell (PCPEC) barrier function and integrity. We now characterized PCPEC cell death and investigated whether apoptosis or necrosis is responsible for the cytotoxicity after infection with different S. suis isolates. We found S. suis strain-dependent histone associated DNA-fragments quantified by ELISA. This response could partially be inhibited by cylcoheximide, cytochalasin D, dexamethasone, herbimycin A, but most effectively by the pan-caspase inhibitor zVAD-fmk. We further detected caspase-3 and -9 activation after infection with all tested S. suis isolates that could also be blocked by zVAD-fmk. However, we found a significantly stronger caspase activity with the protein kinase inhibitor staurosporine. All tested S. suis isolates induced loss of cell viability in PCPEC as shown with the Live/Dead assay, but strain dependent lactate dehydrogenase-release. Both parameters could not be influenced by zVAD-fmk. Immunostaining showed release of high-mobility group box 1 (HMGB1) protein from the nucleus, indicative of necrosis. Transmission electron microscopy showed cell swelling, cytoplasmic vacuolization, loss of membrane integrity, nuclear fermentation but no nuclear condensation, indices for a primarily necrotic cell morphology. Taken together, our findings indicate that S. suis causes cell death in PCPEC by different mechanisms. Although apoptosis may be involved in the process of PCPEC cell death, necrosis seems to be the predominant mechanism. Through inflammation in the choroid plexus during bacterial meningitis, the blood-cerebrospinal fluid barrier function will be compromised.


Subject(s)
Caspases/metabolism , Choroid Plexus/metabolism , Epithelial Cells/metabolism , HMGB1 Protein/metabolism , Streptococcal Infections/metabolism , Streptococcus suis , Animals , Apoptosis , Cell Death/physiology , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , Cells, Cultured , Choroid Plexus/enzymology , Choroid Plexus/pathology , DNA Fragmentation , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/enzymology , Epithelial Cells/pathology , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Microscopy, Electron, Transmission , Streptococcal Infections/enzymology , Streptococcal Infections/pathology , Swine
17.
Cancer Res ; 64(19): 7065-72, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466201

ABSTRACT

We have shown previously that ionizing radiation (IR) induces a persistent G(2)-M arrest but not cell death in MCF-7 breast carcinoma cells that harbor functional p53 but lack caspase-3. In the present study, we investigated the mechanisms of apoptosis resistance and the roles of p53, caspase-3, and cell cycle arrest in IR-induced apoptosis. The methylxanthine caffeine and the staurosporine analog UCN-01, which can inhibit ATM and Chk kinases, efficiently abrogated the IR-induced G(2)-M arrest and induced mitochondrial activation as judged by the loss of the mitochondrial membrane potential and the release of cytochrome c and Smac/Diablo. However, despite these proapoptotic alterations, cell death and activation of the initiator caspase-9 were not induced in MCF-7 cells but were interestingly only observed after reexpression of caspase-3. Sensitization to IR-induced apoptosis by caffeine or UCN-01 was abrogated neither by cycloheximide nor by pifithrin-alpha, an inhibitor of the transcriptional activity of p53. Furthermore, suppression of p53 by RNA interference could not prevent caffeine- and IR-induced mitochondrial alterations and apoptosis but resulted in an even more pronounced G(2)-M arrest. Collectively, our results clearly show that the resistance of MCF-7 cells to IR-induced apoptosis is caused by two independent events; one of them is a caffeine- or UCN-01-inhibitable event that does not depend on p53 or a release of the G(2)-M arrest. The second event is the loss of caspase-3 that surprisingly seems essential for a fully functional caspase-9 pathway, even despite the previous release of mitochondrial proapoptotic proteins.


Subject(s)
Apoptosis/radiation effects , Breast Neoplasms/pathology , Caffeine/pharmacology , Caspases/deficiency , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle/radiation effects , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , G2 Phase/drug effects , G2 Phase/radiation effects , Humans , Mitochondria/drug effects , Mitochondria/radiation effects , Mitosis/drug effects , Mitosis/radiation effects , Radiation Tolerance/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/radiation effects , Transfection , Tumor Suppressor Protein p53/genetics
18.
Oncotarget ; 7(13): 15915-29, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26895377

ABSTRACT

MicroRNAs (miRNAs), a class of small non-coding RNAs that usually cause gene silencing by translational repression or degradation of mRNAs, are implicated in DNA damage-induced stress responses. To identify senescence-associated miRNAs, we performed microarray analyses using wild-type and p53-deficient HCT116 colon carcinoma cells that following gamma-irradiation (γIR) are driven into senescence and apoptosis, respectively. Several miRNAs including miR-30e were found upregulated in a p53-dependent manner specifically in senescent cells, but not in apoptotic cells. Overexpression of miR-30e in HCT116 cells not only inhibited γIR-, etoposide- or miR-34a-induced caspase-3-like DEVDase activities and cell death, but greatly accelerated and augmented their senescent phenotype. Consistently, procaspase-3 protein, but not mRNA decreased in the presence of miR-30e, whereas expression of the cyclin-dependent kinase inhibitor p21 increased both at the mRNA and protein level. Performing luciferase reporter gene assays, we identified the 3'-UTR of the caspase-3 mRNA as a direct miR-30e target. In contrast, although miR-30e was unable to bind to the p21 mRNA, it increased expression of a luciferase construct containing the p21 promoter, suggesting that the miR-30e-mediated upregulation of p21 occurs indirectly at the transcriptional level. Interestingly, despite suppressing procaspase-3 expression, miR-30e was unable to protect RKO colon carcinoma cells from DNA damage-induced death or to induce senescence, as miR-30e completely fails to upregulate p21 in these cells. These data suggest that miR-30e functions in a cell type-dependent manner as an important molecular switch for DNA damage-induced stress responses and may thus represent a target of therapeutic value.


Subject(s)
Caspase 3/biosynthesis , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DNA Damage/physiology , MicroRNAs/metabolism , Gene Expression Regulation , HCT116 Cells , Humans
19.
Oncogene ; 22(39): 8021-30, 2003 Sep 11.
Article in English | MEDLINE | ID: mdl-12970750

ABSTRACT

Cyclooxygenase-2 (COX-2) is involved in diverse processes such as inflammation, carcinogenesis and apoptosis. As COX-2 inhibitors interfere with these processes, inhibition of COX-2 has been suggested as a promising anticancer treatment. However, the role of COX-2 in modulation of apoptosis as well as the death pathways affected by COX-2 inhibitors are poorly characterized. Here we demonstrate that the selective COX-2 inhibitors NS-398 and nimesulide increased TNF sensitivity of TNF-resistant HeLa H21 and TNF-sensitive HeLa D98 cells, although this cytokine induced significant COX-2 activity, as judged by prostaglandin E(2) (PGE(2)) production, only in H21 cells. TNF did also not induce PGE(2) production in MCF-7/casp-3 cells stably expressing COX-2; however, nimesulide strongly enhanced TNF-induced apoptosis in these cells. Furthermore, COX-2 activity in HeLa H21 cells could be inhibited by NS-398 concentrations that were 10 000-fold lower compared to those required for the induction of cell death. Most intriguingly, sensibilization to apoptosis was specifically observed in response to activation of death receptors. Not only TNF-induced cell death but also apoptosis triggered by the CD95 and TRAIL receptors was enhanced by nimesulide. In contrast, apoptosis induced by the anticancer drugs doxorubicine and etoposide that target the mitochondrial death pathway remained unaffected. Together, our data suggest that COX-2 inhibitors overcome apoptosis resistance and selectively sensitize tumor cells to the extrinsic death receptor-induced apoptotic pathway independently of COX-2.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Apoptosis Regulatory Proteins , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3 , Caspases/drug effects , Caspases/genetics , Caspases/metabolism , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/metabolism , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Etoposide/pharmacology , Female , HeLa Cells/drug effects , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Membrane Proteins , Mitochondria/drug effects , Mitochondria/metabolism , Nitrobenzenes/pharmacology , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Tumor Necrosis Factor/drug effects , Sulfonamides/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/immunology , fas Receptor/metabolism
20.
Oncogene ; 22(5): 775-80, 2003 Feb 06.
Article in English | MEDLINE | ID: mdl-12569371

ABSTRACT

Activation of caspases has been demonstrated to be involved in thrombocytopenia and prolonged storage of platelet concentrates. Platelets represent enucleate cells that comprise all elements of the mitochondrial apoptosis pathway. However, no apoptotic stimuli capable of activating the endogenous caspase cascade have been identified so far. Using tributyltin (TBT) we could identify a compound that is capable of activating caspase-9 and -3 in platelets. Recent studies implicate that TBT induces apoptosis via the mitochondrial signaling pathway that is characterized by the formation of a high-molecular-weight complex (apoptosome) containing the adapter protein Apaf-1 and active caspase-9. Interestingly, addition of TBT induced the activation of caspase-9 in an ultra-rapid kinetic within the first 2 min. In addition, size exclusion chromatography revealed that TBT-mediated processing of caspase-9 occurs in the absence of the apoptosome. Thus, these data implicate that TBT induces the activation of caspase-9 by a mechanism not involving the formation of the apoptosome.


Subject(s)
Blood Platelets/drug effects , Caspases/drug effects , Enzyme Activation/drug effects , Trialkyltin Compounds/pharmacology , Apoptosis/drug effects , Caspase 9 , Caspases/metabolism , Enzyme Activation/physiology , Humans , Time Factors , Trialkyltin Compounds/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL