Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nat Immunol ; 17(11): 1282-1290, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27618552

ABSTRACT

Glioma cells recruit and exploit microglia (the resident immune cells of the brain) for their proliferation and invasion ability. The underlying molecular mechanism used by glioma cells to transform microglia into a tumor-supporting phenotype has remained elusive. We found that glioma-induced microglia conversion was coupled to a reduction in the basal activity of microglial caspase-3 and increased S-nitrosylation of mitochondria-associated caspase-3 through inhibition of thioredoxin-2 activity, and that inhibition of caspase-3 regulated microglial tumor-supporting function. Furthermore, we identified the activity of nitric oxide synthase 2 (NOS2, also known as iNOS) originating from the glioma cells as a driving stimulus in the control of microglial caspase-3 activity. Repression of glioma NOS2 expression in vivo led to a reduction in both microglia recruitment and tumor expansion, whereas depletion of microglial caspase-3 gene promoted tumor growth. Our results provide evidence that inhibition of the denitrosylation of S-nitrosylated procaspase-3 mediated by the redox protein Trx2 is a part of the microglial pro-tumoral activation pathway initiated by glioma cancer cells.


Subject(s)
Caspase 3/metabolism , Glioma/metabolism , Glioma/pathology , Microglia/metabolism , Phenotype , Animals , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Enzyme Activation , Gene Knockdown Techniques , Glioma/immunology , Heterografts , Humans , Male , Mice , Microglia/immunology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Thioredoxins/metabolism , Tumor Burden
2.
Proc Natl Acad Sci U S A ; 114(30): E6137-E6146, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696319

ABSTRACT

Neuroblastoma is a pediatric cancer characterized by variable outcomes ranging from spontaneous regression to life-threatening progression. High-risk neuroblastoma patients receive myeloablative chemotherapy with hematopoietic stem-cell transplant followed by adjuvant retinoid differentiation treatment. However, the overall survival remains low; hence, there is an urgent need for alternative therapeutic approaches. One feature of high-risk neuroblastoma is the high level of DNA methylation of putative tumor suppressors. Combining the reversibility of DNA methylation with the differentiation-promoting activity of retinoic acid (RA) could provide an alternative strategy to treat high-risk neuroblastoma. Here we show that treatment with the DNA-demethylating drug 5-Aza-deoxycytidine (AZA) restores high-risk neuroblastoma sensitivity to RA. Combined systemic distribution of AZA and RA impedes tumor growth and prolongs survival. Genome-wide analysis of treated tumors reveals that this combined treatment rapidly induces a HIF2α-associated hypoxia-like transcriptional response followed by an increase in neuronal gene expression and a decrease in cell-cycle gene expression. A small-molecule inhibitor of HIF2α activity diminishes the tumor response to AZA+RA treatment, indicating that the increase in HIF2α levels is a key component in tumor response to AZA+RA. The link between increased HIF2α levels and inhibited tumor growth is reflected in large neuroblastoma patient datasets. Therein, high levels of HIF2α, but not HIF1α, significantly correlate with expression of neuronal differentiation genes and better prognosis but negatively correlate with key features of high-risk tumors, such as MYCN amplification. Thus, contrary to previous studies, our findings indicate an unanticipated tumor-suppressive role for HIF2α in neuroblastoma.


Subject(s)
Azacitidine/analogs & derivatives , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Proliferation/genetics , Genetic Therapy/methods , Neuroblastoma/pathology , Tretinoin/therapeutic use , Animals , Azacitidine/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chemotherapy, Adjuvant , Decitabine , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Mice, Nude
4.
J Cell Mol Med ; 15(8): 1757-68, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20807285

ABSTRACT

The transcription factor p73, a member of the p53 family of proteins, is involved in the regulation of cell cycle progression and apoptosis. Due to alternative promoters and carboxy-terminal splicing, the P73 gene gives rise to a range of different isoforms. Interestingly, a particular increase in expression of the TAp73α isoform has been reported in various tumours. In addition, TAp73α has been shown to inhibit Bax activation and mitochondrial dysfunctions and thereby to confer small cell lung carcinoma (SCLC) cells resistance to drug-induced apoptosis. However, the precise mechanism by which TAp73α exerts its pro-survival effect is yet unclear. Here we report that TAp73α, but not TAp73ß, regulates the expression of inducible Hsp72/HSPA1A. Hsp72 proved to be required for the survival effects of TAp73α as antisense knockdown of Hsp72 resulted in an abolishment of the anti-apoptotic effect of TAp73α in SCLC cells upon Etoposide treatment. Importantly, depletion of Hsp72 allowed activation of Bax, loss of mitochondrial membrane potential and lysosomal membrane permeabilization in SCLC cells even in the presence of TAp73α. Finally, we revealed that TAp73ß counteracts the anti-apoptotic effect of TAp73α by preventing Hsp72 induction. Our results thus provide additional evidence for the potential oncogenic role of TAp73α, and extend the understanding of the mechanism for its anti-apoptotic effect.


Subject(s)
Alternative Splicing , Apoptosis , DNA-Binding Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HSP72 Heat-Shock Proteins/genetics , Humans , Immunoblotting , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Nuclear Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , bcl-2-Associated X Protein/metabolism
5.
J Biol Chem ; 285(5): 3462-9, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19917601

ABSTRACT

Proinsulin C-peptide is internalized into cells, but a function of its intracellular localization has not been established. We now demonstrate that, upon cellular entry, C-peptide is localized to the nucleoli, where it promotes transcription of genes encoding for ribosomal RNA. We find that C-peptide binds to histones and enhances acetylation of lysine residue 16 of histone H4 at the promoter region of genes for ribosomal RNA. In agreement with synchrony of ribosomal RNA synthesis and cell proliferation, we show that C-peptide stimulates proliferation in chondrocytes and HEK-293 cells. This regulation of ribosomal RNA provides a mechanism by which C-peptide can exert transcriptional effects and implies that the peptide has growth factor activity.


Subject(s)
C-Peptide/chemistry , Gene Expression Regulation , RNA, Ribosomal/metabolism , 3T3 Cells , Animals , Cell Cycle , Cell Line , Cell Nucleolus/metabolism , Cell Proliferation , Chondrocytes/metabolism , Epigenesis, Genetic , Histones/chemistry , Humans , Mice , Transcription, Genetic
6.
Stem Cell Res ; 16(1): 1-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26610203

ABSTRACT

The zinc finger transcription factor Zac1 is expressed in dividing progenitors of the nervous system with expression levels negatively controlled by genomic imprinting. To explore the consequences of elevated ZAC1 levels during neurogenesis we overexpressed it in the developing CNS. Increased levels of ZAC1 rapidly promoted upregulation of CDK inhibitors P57 and P27 followed by cell cycle exit. Surprisingly this was accompanied by stalled neuronal differentiation. Genome wide expression analysis of cortical cells overexpressing Zac1 revealed a decrease in neuronal gene expression and an increased expression of imprinted genes, factors regulating mesoderm formation as well as features of differentiated muscle. In addition, we observed a rapid induction of several genes regulating pluripotency. Taken together, our data suggests that expression levels of Zac1 need to be kept under strict control to avoid premature cell cycle exit, disrupted neurogenesis and aberrant expression of non-neuronal genes including pluripotency associated factors.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Reprogramming , Neurogenesis , Transcription Factors/metabolism , Animals , Base Sequence , Body Patterning , Cell Cycle , Cell Differentiation , Cell Proliferation , Chickens , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Genome , HEK293 Cells , Humans , Mice , Models, Biological , Muscles/cytology , Neural Tube/embryology , Neural Tube/metabolism , Neurons/cytology , Pluripotent Stem Cells/metabolism , Protein Binding , SOXB1 Transcription Factors
7.
Neuro Oncol ; 16(2): 241-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24335696

ABSTRACT

BACKGROUND: We have previously shown that the transcriptional coregulator NCoR represses astrocytic differentiation of neural stem cells, suggesting that NCoR could be a plausible target for differentiation therapy of glioblastoma. METHODS: To study a putative role for NCoR in regulating glioblastoma cell characteristics, we used RNA-mediated knockdown followed by analysis of gene expression, proliferation and cell growth, autophagy, invasiveness in vitro, and tumor formation in vitro and in vivo. We further performed chromatin immunoprecipitation of NCoR followed by genome-wide sequencing in the human glioblastoma cell line U87 in order to reveal NCoR-occupied loci. RESULTS: RNA knockdown of NCoR resulted in a moderate increase in differentiation accompanied by a significant decrease in proliferation in adherent U87 human glioblastoma cells. chromatin immunoprecipitation sequencing approach revealed alternative mechanisms underlying the decrease in proliferation, as NCoR was enriched at promoters of genes associated with autophagy such as ULK3. Indeed, signs of an autophagy response in adherent glioblastoma cells included an increased expression of autophagy genes, such as Beclin1, and increased lipidation and nuclear puncta of LC3. Intriguingly, in parallel to the effects in the adherent cells, NCoR knockdown resulted in a significant increase in anchorage-independent growth, and this glioblastoma cell population showed dramatic increases in invasive properties in vitro and tumor formation capacity in vitro and in vivo along with an increased proliferation rate. CONCLUSION: Our results unveil unexpected aspects of NCoR regulation of tumor characteristics in glioblastoma cells and highlight the need for caution when transposing developmental concepts directly to cancer therapy.


Subject(s)
Autophagy , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Glioblastoma/pathology , Nuclear Receptor Co-Repressor 1/metabolism , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Chromatin Immunoprecipitation , Female , Flow Cytometry , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Receptor Co-Repressor 1/antagonists & inhibitors , Nuclear Receptor Co-Repressor 1/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Dev Cell ; 26(3): 223-36, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23948251

ABSTRACT

The chromatin remodeler CHD5 is expressed in neural tissue and is frequently deleted in aggressive neuroblastoma. Very little is known about the function of CHD5 in the nervous system or its mechanism of action. Here we report that depletion of Chd5 in the developing neocortex blocks neuronal differentiation and leads to an accumulation of undifferentiated progenitors. CHD5 binds a large cohort of genes and is required for facilitating the activation of neuronal genes. It also binds a cohort of Polycomb targets and is required for the maintenance of H3K27me3 on these genes. Interestingly, the chromodomains of CHD5 directly bind H3K27me3 and are required for neuronal differentiation. In the absence of CHD5, a subgroup of Polycomb-repressed genes becomes aberrantly expressed. These findings provide insights into the regulatory role of CHD5 during neurogenesis and suggest how inactivation of this candidate tumor suppressor might contribute to neuroblastoma.


Subject(s)
DNA Helicases/physiology , Nerve Tissue Proteins/physiology , Neuroblastoma/genetics , Neurogenesis/genetics , Neurons/cytology , Polycomb-Group Proteins/genetics , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Embryonic Stem Cells/cytology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred Strains , Neuroblastoma/pathology , Pregnancy , Retina/cytology
9.
Oncotarget ; 2(12): 1145-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22201672

ABSTRACT

Caspase-2 is ubiquitously expressed and the most evolutionarily conserved mammalian caspase. It can be activated by a range of death stimuli prior to Bax activation and the occurrence of apoptotic mitochondrial dysfunctions. Caspase-2 has also been reported to exert tumour suppressor function in vivo. The full length TAp73alpha isoform is found up-regulated in various tumour types, and is reported in a cell-type specific manner to repress drug-induced apoptosis. Here, we report that TAp73alpha represses caspase-2 enzymatic activity and by this means reduce caspase-2 induced Bax activation, loss of mitochondrial transmembrane potential and resulting apoptosis. The inhibitory effect on caspase-2 requires the presence of the DNA binding domain and SAM domain region of TAp73alpha. In conclusion, the ability of TAp73alpha to act as an inhibitor of caspase-2-induced cell death together with its up-regulation in certain tumour types strengthen the potential oncogenic activities for this protein.


Subject(s)
Apoptosis/genetics , Caspase 2/metabolism , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/metabolism , Lung Neoplasms/metabolism , Mitochondria/metabolism , Nuclear Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Tumor Suppressor Proteins/metabolism , Caspase 2/genetics , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Enzyme Activation , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial , Mitochondria/physiology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Tumor Protein p73 , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
10.
Mol Cell Biol ; 29(7): 1814-25, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19158275

ABSTRACT

The transcription factor p73, a member of the p53 family of proteins, is involved in the regulation of cell cycle progression and apoptosis. However, the regulatory mechanisms controlling the distinct roles for p73 in these two processes have remained unclear. Here, we report that p73 is able to induce cell cycle arrest independently of its amino-terminal transactivation domain, whereas this domain is crucial for p73 proapoptotic functions. We also characterized a second transactivation domain in the carboxy terminus of p73 within amino acid residues 381 to 399. This carboxy terminus transactivation domain was found to preferentially regulate genes involved in cell cycle progression. Moreover, its activity is regulated throughout the cell cycle and modified by protein kinase C-dependent phosphorylation at serine residue 388. Our results suggest that this novel posttranslational modification within the p73 carboxy terminus transactivation domain is involved in the context-specific guidance of p73 toward the selective induction of cell cycle arrest.


Subject(s)
Cell Cycle , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Kinase C/metabolism , Transcriptional Activation/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Herpes Simplex Virus Protein Vmw65/metabolism , Humans , Molecular Sequence Data , Mutant Proteins/metabolism , Organ Specificity , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Serine/metabolism , Structure-Activity Relationship
11.
Mol Cancer Ther ; 8(12): 3307-17, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19952118

ABSTRACT

Lamellarin D, a potent cytotoxic marine alkaloid, exerts its antitumor action through two complementary pathways: a nuclear route via topoisomerase I inhibition and a mitochondrial targeting. The present study was designed to investigate the contribution of these two pathways for apoptosis in cancer cells. Lamellarin D promoted nuclear apoptosis in leukemia cells without prominent cell cycle arrest. Signals transmitted by lamellarin D initiated apoptosis via the intrinsic apoptotic pathway. The drug induced conformational activation of Bax and decreased the expression levels of antiapoptotic proteins Bcl-2 and cIAP2 in association with activation of caspase-9 and caspase-3. Upon lamellarin D exposure, Fas and Fas-L expression was not modified in leukemia cells. Moreover, leukemia cells deficient in caspase-8 or Fas-associated protein with death domain underwent apoptosis through the typical mitochondrial apoptotic cascade, indicating that cell death induced by lamellarin D was independent of the extrinsic apoptotic pathway. Lamellarin D also exerted a topoisomerase I-mediated DNA damage response resulting in H2AX phosphorylation, and the upregulation of the DNA repair protein Rad51 and of p53, as well as the phosphorylation of p53 at serine 15. However, lamellarin D killed efficiently mutated p53 or p53 null cancer cells, and sensitivity to lamellarin D was abrogated neither by cycloheximide nor in enucleated cells. Lamellarin D-induced cytochrome c release occurs independently of nuclear factors in a cell-free system. These results suggest that lamellarin D exerts its cytotoxic effects primarily by inducing mitochondrial apoptosis independently of nuclear signaling. Thus, lamellarin D constitutes a new proapoptotic agent that may bypass certain forms of apoptosis resistance that occur in tumor cells.


Subject(s)
Apoptosis/drug effects , Coumarins/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Isoquinolines/pharmacology , Mitochondria/metabolism , Animals , Apoptosis/physiology , Baculoviral IAP Repeat-Containing 3 Protein , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , DNA Damage , DNA Topoisomerases, Type I/metabolism , Flow Cytometry , HCT116 Cells , Histones/metabolism , Humans , Immunoblotting , Inhibitor of Apoptosis Proteins/metabolism , Jurkat Cells , Microscopy, Fluorescence , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/physiopathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases , bcl-2-Associated X Protein/metabolism
12.
PLoS One ; 4(9): e7226, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19789652

ABSTRACT

BACKGROUND: Electrical pulses have been used to enhance uptake of molecules into living cells for decades. This technique, often referred to as electroporation, has become an increasingly popular method to enhance in vivo DNA delivery for both gene therapy applications as well as for delivery of vaccines against both infectious diseases and cancer. In vivo electrovaccination (gene delivery followed by electroporation) is currently being investigated in several clinical trials, including DNA delivery to healthy volunteers. However, the mode of action at molecular level is not yet fully understood. METHODOLOGY/PRINCIPAL FINDINGS: This study investigates intradermal DNA electrovaccination in detail and describes the effects on expression of the vaccine antigen, plasmid persistence and the local tissue environment. Gene profiling of the vaccination site showed that the combination of DNA and electroporation induced a significant up-regulation of pro-inflammatory genes. In vivo imaging of luciferase activity after electrovaccination demonstrated a rapid onset (minutes) and a long duration (months) of transgene expression. However, when the more immunogenic prostate specific antigen (PSA) was co-administered, PSA-specific T cells were induced and concurrently the luciferase expression became undetectable. Electroporation did not affect the long-term persistence of the PSA-expressing plasmid. CONCLUSIONS/SIGNIFICANCE: This study provides important insights to how DNA delivery by intradermal electrovaccination affects the local immunological responses of the skin, transgene expression and clearance of the plasmid. As the described vaccination approach is currently being evaluated in clinical trials, the data provided will be of high significance.


Subject(s)
DNA/metabolism , Injections, Intradermal/methods , Skin/metabolism , Transgenes , Animals , Electrophysiology/methods , Electroporation , Female , Gene Transfer Techniques , Genetic Therapy/methods , Kinetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmids/metabolism
13.
J Biol Chem ; 280(40): 34159-69, 2005 Oct 07.
Article in English | MEDLINE | ID: mdl-16087678

ABSTRACT

The p73 gene, a member of the p53 family, encodes several variants through differential splicing and use of alternative promoters. At the NH2 terminus, two different promoters generate the full-length and the DeltaN isoforms, with or without the transactivating domain. At the COOH terminus, seven isoforms generated through alternative splicing have been cloned. Previous studies have demonstrated that DeltaNp73 isoforms exert a dominant-negative effect on p73 by blocking their transactivation activity and hence the ability to induce apoptosis. Considerable efforts are made to identify the functional diversity of the COOH-terminal p73 variants. In this study, we found that p73alpha inhibited drug-induced apoptosis in small cell lung carcinoma cells, whereas p73beta promoted it. p73alpha prevented Bax activation, mitochondrial dysfunction, and caspase activation. In addition, p73alpha was also able to reduce apoptosis induced by the BH3-only protein PUMA (p53 up-regulated modulator of apoptosis). Furthermore, we discovered that p73alpha is able to inhibit the pro-apoptotic effect of p73beta, demonstrating the existence of equilibrium between these two p73 isoforms. In conclusion, the reported overexpression of p73alpha in certain tumor types, and our findings that p73alpha exerts anti-apoptotic functions, indicate a potential oncogenic activity for p73.


Subject(s)
Apoptosis/genetics , Carcinoma, Small Cell/genetics , DNA-Binding Proteins/physiology , Genes, Tumor Suppressor/physiology , Lung Neoplasms/genetics , Nuclear Proteins/physiology , Alternative Splicing , Apoptosis Regulatory Proteins/physiology , Carcinoma, Small Cell/pathology , Gene Expression Profiling , Humans , Lung Neoplasms/pathology , Protein Isoforms , Proto-Oncogene Proteins/physiology , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL