Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Sci Rep ; 10(1): 8286, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32427867

ABSTRACT

The posttranslational modification ADP-ribosylation is involved in many cellular processes, with distinct roles for poly- and mono(ADP-ribosyl)ation (PAR- and MARylation, respectively). Reversibility of intracellular MARylation was demonstrated with the discovery of MACROD1, MACROD2 and TARG1, three macrodomain-containing enzymes capable of reversing MARylation of proteins and RNA. While the three enzymes have identical activities in vitro, their roles in cells are unclear and published data are partially contradictory, possibly due to a lack of validated reagents. We developed monoclonal antibodies to study these proteins and analysed their tissue distribution and intracellular localisation. MACROD1 is most prevalent in mitochondria of skeletal muscle, MACROD2 localises to nucleo- and cytoplasm and is found so far only in neuroblastoma cells, whereas the more ubiquitously expressed TARG1 is present in nucleoplasm, nucleolus and stress granules. Loss of MACROD1 or loss of TARG1 leads to disruption of mitochondrial or nucleolar morphology, respectively, hinting at their importance for these organelles. To start elucidating the underlying mechanisms, we have mapped their interactomes using BioID. The cellular localisation of interactors supports the mitochondrial, nucleolar and stress granule localisation of MACROD1 and TARG1, respectively. Gene ontology analysis suggests an involvement of MACROD1 and TARG1 in RNA metabolism in their respective compartments. The detailed description of the hydrolases' expression, localisation and interactome presented here provides a solid basis for future work addressing their physiological function in more detail.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Cell Line , Cell Nucleolus/metabolism , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Jurkat Cells , MCF-7 Cells , Mitochondria, Muscle/metabolism , Protein Binding , Tissue Distribution
2.
J Eur Acad Dermatol Venereol ; 33(2): 367-375, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30357932

ABSTRACT

BACKGROUND: Janus kinase (JAK) inhibition may be a promising new treatment modality for inflammatory (skin) diseases. However, little is known about direct effects of kinase inhibitors on keratinocyte differentiation and function as well as skin barrier formation. OBJECTIVE: Our aim was to address the direct impact of kinase inhibition of the JAK1/3 pathways by tofacitinib on keratinocyte immune function and barrier formation in atopic dermatitis (AD) and psoriasis. METHODS: 3D skin equivalents of both diseases were developed and concurrently pretreated with tofacitinib. To induce AD, 3D skin equivalents were stimulated with recombinant human IL-4 and IL-13. Psoriasis-like conditions were induced by incubation with IL-17A, IL-22 and tumour necrosis factor α (TNFα). The activation of signal transducer and activator of transcription (STAT)1, STAT3 and STAT6 was assessed by Western blot analysis. Microarray analysis and quantitative real-time PCR were used for gene expression analysis. RESULTS: Tofacitinib pretreatment preserved epidermal morphology and reduced STAT3 and STAT6 phosphorylation of AD-like and STAT3 phosphorylation of psoriasis-like culture conditions in 3D skin models compared to sham-controls. Filaggrin expression was fully maintained in the AD-like models, but only partially in psoriasis-like conditions after pretreatment with tofacitinib. In addition, tofacitinib upregulated DSC1, FLG and KRT1. Using gene expression analysis, downregulation of POSTN and IL24 was observed in AD-like conditions, whereas downregulation of IL20 and IL1B was observed in psoriasis-like conditions. CONCLUSION: JAK1/3 inhibition counteracted cytokine-induced AD- and psoriasis-like epidermal morphology and enhanced keratinocyte differentiation in 3D skin models. This effect was more pronounced in the AD-like models compared to the psoriasis-like 3D skin models.


Subject(s)
Dermatitis, Atopic/pathology , Imaging, Three-Dimensional , Intermediate Filament Proteins/pharmacology , Janus Kinase 1/drug effects , Piperidines/pharmacology , Psoriasis/pathology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Cell Proliferation/drug effects , Computer Simulation , Dermatitis, Atopic/drug therapy , Filaggrin Proteins , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Psoriasis/drug therapy , STAT6 Transcription Factor/drug effects , Sensitivity and Specificity
4.
Mol Psychiatry ; 22(6): 920-930, 2017 06.
Article in English | MEDLINE | ID: mdl-27821870

ABSTRACT

Major depressive disorder (MDD) is associated with reduced concentrations of γ-aminobutyric acid (GABA) that are normalized by antidepressant therapies. Moreover, depressive-like phenotypes of GABAA receptor mutant mice can be reversed by treatment with conventional antidepressants drugs, as well as by subanesthetic doses of ketamine. Thus GABAergic deficits may causally contribute to depressive disorders, while antidepressant therapies may enhance GABAergic synaptic transmission. Here we tested the hypothesis that sustained enhancement of GABAergic transmission alone is sufficient to elicit antidepressant-like behavior, using disinhibition of GABAergic interneurons. We focused on somatostatin-positive (SST+) GABAergic interneurons because of evidence that their function is compromised in MDD. To disinhibit SST+ interneurons, we inactivated the γ2 subunit gene of GABAA receptors selectively in these neurons (SSTCre:γ2f/f mice). Loss of inhibitory synaptic input resulted in increased excitability of SST+ interneurons. In turn, pyramidal cell targets of SST+ neurons showed an increased frequency of spontaneous inhibitory postsynaptic currents. The behavior of SSTCre:γ2f/f mice mimicked the effects of anxiolytic and antidepressant drugs in a number of behavioral tests, without affecting performance in a spatial learning- and memory-dependent task. Finally, brain extracts of SSTCre:γ2f/f mice showed decreased phosphorylation of the eukaryotic elongation factor eEF2, reminiscent of the effects of ketamine. Importantly, these effects occurred without altered activity of the mammalian target of rapamycin pathway nor did they involve altered expression of SST. However, they were associated with reduced Ca2+/calmodulin-dependent auto-phosphorylation of eEF2 kinase, which controls the activity of eEF2 as its single target. Thus enhancing GABAergic inhibitory synaptic inputs from SST+ interneurons to pyramidal cells and corresponding chronic reductions in the synaptic excitation:inhibition ratio represents a novel strategy for antidepressant therapies that reproduces behavioral and biochemical end points of rapidly acting antidepressants.


Subject(s)
GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Inhibitory Postsynaptic Potentials/physiology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain/metabolism , Depressive Disorder, Major/drug therapy , GABA Agents/metabolism , GABA Agents/therapeutic use , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/physiology , Ketamine/pharmacology , Mice , Mice, Transgenic , Receptors, GABA-A/metabolism , Somatostatin/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
6.
Br J Dermatol ; 165(5): 966-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21711332

ABSTRACT

BACKGROUND: Interleukin (IL)-31 is a novel Th2 T-cell cytokine that induces pruritus and dermatitis in transgenic mice. While enhanced mRNA expression of this cytokine is detected in skin samples of inflammatory skin diseases, the regulation of IL-31 expression is poorly understood. OBJECTIVES: To assess the effects of ultraviolet (UV) B radiation and H2O2 on IL-31 mRNA and protein expression in skin and different peripheral blood mononuclear cells (PBMCs). METHODS: The effects of UVB radiation and H2O2, as a prototypic reactive oxygen species, on IL-31 mRNA and protein expression were analysed in various inflammation-related cells and murine skin tissue. RESULTSTreatment of cells with UVB radiation and H2 O2 strongly induced IL-31 mRNA and protein expression in human PBMCs and in the skin of SKH-1 mice. Following exposure to UVB or H2O2, we observed increased expression of IL-31 mRNA in T cells, monocytes, macrophages, and immature and especially mature dendritic cells. H2O2 treatment but not UVB radiation led to a moderate upregulation of IL-31 mRNA expression in epidermal keratinocytes and dermal fibroblasts. Pretreatment of T lymphocytes with the MAPK p38 inhibitor SB203580 or the MEK1 inhibitor U0126 reduced the stimulatory effect of H2O2. These experiments suggest that p38 is involved in the regulation of IL-31 expression in human skin. CONCLUSIONS: Our studies reveal that UVB and reactive oxygen species stimulate the expression of IL-31 in PBMCs and skin, especially in T cells, monocytes and monocyte-derived dendritic cells.


Subject(s)
Dendritic Cells/radiation effects , Hydrogen Peroxide/pharmacology , Interleukins/metabolism , Leukocytes, Mononuclear/radiation effects , Reactive Oxygen Species/pharmacology , T-Lymphocytes/radiation effects , Animals , Cells, Cultured , Fibroblasts/metabolism , Humans , Keratinocytes/metabolism , Mice , Mice, Hairless , RNA, Messenger/metabolism , Skin/metabolism , Ultraviolet Rays
7.
Mol Psychiatry ; 16(4): 383-406, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21079608

ABSTRACT

Increasing evidence points to an association between major depressive disorders (MDDs) and diverse types of GABAergic deficits. In this review, we summarize clinical and preclinical evidence supporting a central and causal role of GABAergic deficits in the etiology of depressive disorders. Studies of depressed patients indicate that MDDs are accompanied by reduced brain concentration of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) and by alterations in the subunit composition of the principal receptors (GABA(A) receptors) mediating GABAergic inhibition. In addition, there is abundant evidence that suggests that GABA has a prominent role in the brain control of stress, the most important vulnerability factor in mood disorders. Furthermore, preclinical evidence suggests that currently used antidepressant drugs (ADs) designed to alter monoaminergic transmission and nonpharmacological therapies may ultimately act to counteract GABAergic deficits. In particular, GABAergic transmission has an important role in the control of hippocampal neurogenesis and neural maturation, which are now established as cellular substrates of most if not all antidepressant therapies. Finally, comparatively modest deficits in GABAergic transmission in GABA(A) receptor-deficient mice are sufficient to cause behavioral, cognitive, neuroanatomical and neuroendocrine phenotypes, as well as AD response characteristics expected of an animal model of MDD. The GABAergic hypothesis of MDD suggests that alterations in GABAergic transmission represent fundamentally important aspects of the etiological sequelae of MDDs that are reversed by monoaminergic AD action.


Subject(s)
Depressive Disorder, Major/genetics , Genetic Predisposition to Disease , gamma-Aminobutyric Acid/deficiency , gamma-Aminobutyric Acid/genetics , Animals , Antidepressive Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mutation/genetics , Receptors, GABA/deficiency , Receptors, GABA/genetics , Risk Factors
8.
Cell Mol Life Sci ; 65(20): 3255-64, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18636226

ABSTRACT

Mitogenic signals stimulate cell division by activating cyclin/cyclin-dependent kinase (CDK) complexes. Their timely regulation ensures proper cell cycle progression. It is therefore not surprising that cyclin/CDK complexes are integrators of multiple signals from both the extracellular environment and intracellular cues. Important regulators of cyclin/CDKs are the CDK inhibitors that have attracted attention due to their association with disease. p27(KIP1) is a CDK inhibitor that controls CDK activity throughout the cell cycle. As a CDK inhibitor, p27(KIP1) has tumor suppressor activity. Besides CDKs, p27(KIP1) regulates additional cellular processes, including cell motility, some of which seem to mediate oncogenic activities of p27(KIP1). These activities of p27(KIP1) are regulated through multiple phosphorylation sites, targeted by several signal transduction pathways. Understanding functions and regulation of p27(KIP1) will be important to determine which isoform of p27(KIP1) has anti- or pro-tumorigenic activities. Such knowledge might be of prognostic value and may offer novel therapeutic windows.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Protein Processing, Post-Translational , Animals , Cell Cycle/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Humans , Mitogens/pharmacology , Neoplasms/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational/drug effects
9.
Oncogene ; 26(35): 5046-59, 2007 Aug 02.
Article in English | MEDLINE | ID: mdl-17310986

ABSTRACT

The phosphoinositide-3-kinase (PI3K)/Akt signaling pathway plays an important role in cell survival and the development of cancer. Macrophage migration inhibitory factor (MIF) is a critical inflammatory cytokine that was recently associated with tumorigenesis and that potently inhibits apoptosis. This may involve inhibition of p53-dependent genes, but the initiating molecular mechanism of how MIF controls survival/apoptosis is unknown. Here, we show that MIF prevents apoptosis and promotes tumor cell survival by directly activating the Akt pathway. MIF enhanced Akt activity in primary and immortalized fibroblasts (MEF and NIH/3T3), HeLa cervix carcinoma cells and various breast cancer cell lines. Activation was abolished by kinase inhibitors Ly294002 and PP2 and in Src/Yes/Fyn(SYF)(-/-) and CD74(-/-)(MEFs), while being enhanced in CD74-overexpressing MEFs, demonstrating that the MIF-induced Akt pathway encompasses signaling through the MIF receptor CD74 and the upstream kinases Src and PI3K. Akt was activated by exogenous rMIF and autocrine MIF action, as revealed by experiments in MIF(-/-)MEFs and antibody blockade. siRNA knockdown of CSN5/JAB1, a tumor marker and MIF-binding protein, showed that JAB1 controls autocrine MIF-mediated Akt signaling by inhibition of MIF secretion. Akt activation by MIF led to phosphorylation of the proapoptotic proteins BAD and Foxo3a. Apoptosis inhibition by MIF was functionally associated with Akt activation as it was abolished by overexpression of the Akt pathway inhibitor PTEN and occurred independently of p53. This was shown by studying DNA damage-induced apoptosis in fibroblasts, the Fas death pathway in HeLa cells that do not express functional p53, and etoposide-induced apoptosis in breast carcinoma cells expressing mutant p53. Importantly, dependence of breast cancer cell survival on MIF correlated with Akt activation and the PTEN status of these cells. Thus, MIF can directly promote cell survival through activation of the PI3K/Akt pathway and this effect is critical for tumor cell survival.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Macrophage Migration-Inhibitory Factors/metabolism , Neoplasms/metabolism , Peptide Hydrolases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Autocrine Communication , Breast Neoplasms/metabolism , COP9 Signalosome Complex , Cell Line, Tumor , Cell Survival , Histocompatibility Antigens Class II/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Mice , Neoplasms/pathology , Peptide Hydrolases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , src-Family Kinases/metabolism
10.
Curr Top Microbiol Immunol ; 302: 63-122, 2006.
Article in English | MEDLINE | ID: mdl-16620026

ABSTRACT

A significant body of evidence has been accumulated that demonstrates decisive roles of members of the Myc/Max/Mad network in the control of various aspects of cell behavior, including proliferation, differentiation, and apoptosis. The components of this network serve as transcriptional regulators. Mad family members, including Mad1, Mxi1, Mad3, Mad4, Mnt, and Mga, function in part as antagonists of Myc oncoproteins. At the molecular level this antagonism is reflected by the different cofactor/chromatin remodeling complexes that are recruited by Myc and Mad family members. One important function of the latter is their ability to repress gene transcription. In this review we summarize the current view of how this repression is achieved and what the consequences of Mad action are for cell behavior. In addition, we point out some of the many aspects that have not been clarified and thus leave us with a rather incomplete picture of the functions, both molecular and at the cellular level, of Mad family members.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Humans , Models, Biological , Molecular Sequence Data , Molecular Structure , Multiprotein Complexes , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sequence Homology, Amino Acid
11.
Biochem Soc Trans ; 31(Pt 4): 889-92, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12887328

ABSTRACT

Postsynaptic clustering of GABAA (type A gamma-aminobutyric acid) receptors is essential to ensure proper function of GABAergic synapses. This process is initiated during synapse formation and is maintained throughout life. The tubulin-associated protein gephyrin is required for clustering of GABAA receptors, but its specific role in this process is not understood. A second protein associated selectively with GABAA receptors at postsynaptic sites is dystrophin. It is present in a subset of GABAergic synapses along with several partners, forming the dystrophin-associated protein complex. In this review, we discuss recent advances in the role of neuronal activity and trans-synaptic signaling for the clustering of gephyrin and dystrophin during synaptogenesis and on the role of these proteins for plasticity and maintenance of mature synapses.


Subject(s)
Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Carrier Proteins/physiology , Dystrophin/chemistry , Dystrophin/metabolism , Membrane Proteins/physiology , Neurons/metabolism , Presynaptic Terminals/metabolism
12.
Gut ; 51(5): 685-90, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12377807

ABSTRACT

BACKGROUND: Damage to the gastrointestinal mucosa results in the acute up-regulation of the trefoil factor family peptides TFF1, TFF2, and TFF3. They possess protective, healing, and tumour suppressive functions. Little is known about the regulation of TFF gene expression. The promoters of all three TFF genes contain binding sites (E box) for upstream stimulating factor (USF) and Myc/Max/Mad network proteins. AIMS: To determine the nature and function of transcription factors that bind to these E boxes and to understand their role for TFF gene expression. METHODS: TFF promoter activities were determined by reporter gene assays. DNA binding was monitored by electromobility shift assays and by chromatin immunoprecipitation analyses. Expression of endogenous TFF was determined by multiplex RT-PCR. RESULTS: It was observed that the TFF2 promoter is specifically and efficiently activated by USF transcription factors but not by c-Myc. USF displayed comparable binding to a high affinity Myc/Max binding site compared with the three TFF E boxes, while c-Myc exhibited lower affinity to the TFF E boxes. In contrast, pronounced binding differences were observed in cells with a strong preference for USF to interact specifically with the TFF2 E box, while Myc was not above background. Exogenous expression of USF was sufficient to activate the chromosomal TFF2 and to a lesser extent, the TFF1 gene. CONCLUSION: These findings define USF factors as regulators of the TFF2 gene and suggest that promoter specific effects are important for a pronounced gene activation of this cytoprotective peptide.


Subject(s)
DNA-Binding Proteins , Gastrointestinal Neoplasms/genetics , Gene Expression Regulation , Growth Substances/genetics , Mucins , Muscle Proteins , Neuropeptides , Peptides/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Binding Sites , Electrophoretic Mobility Shift Assay , Gastrointestinal Neoplasms/metabolism , Growth Substances/metabolism , Humans , Peptides/metabolism , Precipitin Tests , Proto-Oncogene Proteins c-myc/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Trefoil Factor-2 , Trefoil Factor-3 , Tumor Cells, Cultured , Upstream Stimulatory Factors
13.
Oncogene ; 20(51): 7494-504, 2001 Nov 08.
Article in English | MEDLINE | ID: mdl-11709721

ABSTRACT

Activin A, a member of the transforming growth factor beta (TGF-beta) superfamily, affects keratinocyte proliferation and differentiation in vitro and in vivo. However, little is known about the mechanisms of activin action in keratinocytes, and its target genes have not been identified. In this study, we demonstrate that activin A and TGF-beta1 directly induce the expression and activity of Mad1, an antagonist of the c-Myc transcription factor, in the human HaCaT keratinocyte cell line. Expression and activity of Mad1 was strongly induced by both factors in keratinocytes, although the intensity of induction was different for activin A and TGF-beta1. To determine a possible role of activin and TGF-beta in the regulation of mad1 expression in vivo, we analysed its expression during cutaneous wound repair when high levels of these factors are present. Expression of mad1 mRNA and protein, but not of other mad genes, increased significantly after skin injury, particularly in polymorphonuclear leukocytes and in suprabasal keratinocytes of the hyperproliferative epithelium. Elevated levels of mad1 mRNA were also detected in the hyperthickened epidermis of psoriatic patients. Since Mad1 regulates proliferation and/or differentiation of various cell types, our results suggest that this transcription factor mediates at least in the part the anti-mitotic and/or differentiation-inducing activities of TGF-beta and activin in keratinocytes.


Subject(s)
Activins/metabolism , Keratinocytes/metabolism , Phosphoproteins/metabolism , Psoriasis/metabolism , Repressor Proteins/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing , Adult , Aged , Animals , Blotting, Western , COS Cells , Cell Cycle Proteins , Cell Differentiation , Cell Division , Cell Line , Cell Nucleus/metabolism , DNA/metabolism , DNA, Complementary/metabolism , Epidermal Cells , Epidermis/metabolism , Humans , In Situ Hybridization , Mice , Mice, Inbred BALB C , Middle Aged , Neutrophils/metabolism , Nuclear Proteins , Phosphoproteins/physiology , RNA, Messenger/metabolism , Repressor Proteins/physiology , Skin/metabolism , Skin/pathology , Time Factors , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
14.
EMBO Rep ; 2(12): 1125-32, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11743027

ABSTRACT

Upregulation of the proto-oncoprotein Myc, a basic, helix-loop-helix, leucin zipper domain transcription factor has profound consequences on cell proliferation, cell growth and apoptosis. Cell cultures of somatic c-myc-/- rat fibroblasts show extremely prolonged doubling times of 52 h. Using time-lapse microscopy, we show here that individual c-myc-/- cells proceeded within approximately 24 h through the cell cycle as fast as c-myc+/+ cells. However, c-myc-/- cells were highly sensitive to contact inhibition and readily arrested in the cell cycle already at low density. Activation of conditional MycER overcame cell cycle arrest in c-myc-/- cells and led to continuous proliferation at the expense of increased apoptosis at high cell density. Conditional expression of Mad1, a Myc antagonist, represses proliferation of different cell types including U2OS cells. In analogy to the effect of Myc, this occurs mainly by reducing the probability of cells remaining in the cycle. Our data demonstrate that the Myc/Max/Mad network does not regulate the duration of the cell cycle, but the decision of cells to enter or exit the cell cycle.


Subject(s)
Cell Cycle , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Transcription Factors , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Cell Count , Cell Cycle Proteins , Cell Division , Cell Line , Cells, Cultured , Contact Inhibition/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation , Microscopy, Video , Nuclear Proteins , Phosphoproteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Rats , Repressor Proteins/genetics , Time Factors
15.
Oncogene ; 20(50): 7420-4, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11704872

ABSTRACT

Oncogenic activation of c-myb by retroviral insertion has been implicated in tumor formation in chicken and mice. These genetic alterations result in deregulated expression of the c-myb gene and frequently in N-terminal truncation of the c-Myb protein. We demonstrate that truncation of the c-Myb N-terminus affects DNA binding and reporter activation. However, all three mutants, Myb Delta N20, Myb Delta N47 and Myb Delta N71 cooperated with C/EBP beta in reporter assays. In contrast to Myb Delta N20 and Myb Delta N47, however, the Myb Delta N71 mutant failed to activate the chromatin embedded endogenous mim-1 gene together with C/EBP beta. This suggests that an N-terminal region (amino acids 47-71) within repeat 1 (R1) of the murine c-Myb DNA binding domain affects activation of chromosomal target genes in collaboration with C/EBP beta.


Subject(s)
Acetyltransferases , CCAAT-Enhancer-Binding Protein-beta/metabolism , DNA/metabolism , Genes, myb , Proto-Oncogene Proteins c-myb/metabolism , Animals , COS Cells , Chickens , Chlorocebus aethiops , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Macromolecular Substances , Mice , Promoter Regions, Genetic , Protein Binding , Proteins/genetics , Proto-Oncogene Proteins c-myb/chemistry , Recombinant Fusion Proteins/biosynthesis , Sequence Deletion , Transcription Factors/metabolism , Transcriptional Activation
17.
Gene ; 277(1-2): 1-14, 2001 Oct 17.
Article in English | MEDLINE | ID: mdl-11602341

ABSTRACT

The members of the Myc/Max/Mad network function as transcriptional regulators. Substantial evidence has been accumulated over the last years that support the model that Myc/Max/Mad proteins affect different aspects of cell behavior, including proliferation, differentiation, and apoptosis, by modulating distinct target genes. The unbalanced expression of these genes, e.g. in response to deregulated Myc expression, is most likely an important aspect of Myc's ability to stimulate tumor formation. Myc and Mad proteins affect target gene expression by recruiting chromatin remodeling activities. In particular Myc interacts with a SWI/SNF-like complex that may contain ATPase activity. In addition Myc binds to TRRAP complexes that possess histone acetyl transferase activity. Mad proteins, that antagonize Myc function, recruit an mSin3 repressor complex with histone deacetylase activity. Thus the antagonism of Myc and Mad proteins is explained at the molecular level by the recruitment of opposing chromatin remodeling activities.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Repressor Proteins , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Cell Cycle/genetics , Cell Cycle/physiology , DNA-Binding Proteins/genetics , Humans , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction , Transcription Factors/genetics
18.
Biochem J ; 358(Pt 3): 705-15, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11535131

ABSTRACT

Max is the central component of the Myc/Max/Mad network of transcription factors that regulate growth, differentiation and apoptosis. Whereas the Myc and Mad genes and proteins are highly regulated, Max expression is constitutive and no post-translational regulation is known. We have found that Max is targeted during Fas-induced apoptosis. Max is first dephosphorylated and subsequently cleaved by caspases. Two specific cleavage sites for caspases in Max were identified, one at IEVE(10) decreasing S and one at SAFD(135) decreasing G near the C-terminus, which are cleaved in vitro by caspase-5 and caspase-7 respectively. Mutational analysis indicates that both sites are also used in vivo. Thus Max represents the first caspase-5 substrate. The unusual cleavage after a glutamic acid residue is observed only with full-length, DNA-binding competent Max protein but not with corresponding peptides, suggesting that structural determinants might be important for this activity. Furthermore, cleavage by caspase-5 is inhibited by the protein kinase CK2-mediated phosphorylation of Max at Ser-11, a previously mapped phosphorylation site in vivo. These findings suggest that Fas-mediated dephosphorylation of Max is required for cleavage by caspase-5. The modifications that occur on Max in response to Fas signalling affect the DNA-binding activity of Max/Max homodimers. Taken together, our findings uncover three distinct processes, namely dephosphorylation and cleavage by caspase-5 and caspase-7, that target Max during Fas-mediated apoptosis, suggesting the regulation of the Myc/Max/Mad network through its central component.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , DNA-Binding Proteins/metabolism , Glutamic Acid , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , COS Cells , Caspase 7 , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , DNA-Binding Proteins/chemistry , Dimerization , Humans , Immunoglobulin M/pharmacology , Jurkat Cells , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Mapping , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Transcription Factors/chemistry , Transcription Factors/metabolism , Transfection , fas Receptor/physiology
19.
Genes Dev ; 15(16): 2042-7, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11511535

ABSTRACT

Myc oncoproteins promote cell cycle progression in part through the transcriptional up-regulation of the cyclin D2 gene. We now show that Myc is bound to the cyclin D2 promoter in vivo. Binding of Myc induces cyclin D2 expression and histone acetylation at a single nucleosome in a MycBoxII/TRRAP-dependent manner. Down-regulation of cyclin D2 mRNA expression in differentiating HL60 cells is preceded by a switch of promoter occupancy from Myc/Max to Mad/Max complexes, loss of TRRAP binding, increased HDAC1 binding, and histone deacetylation. Thus, recruitment of TRRAP and regulation of histone acetylation are critical for transcriptional activation by Myc.


Subject(s)
Bacterial Proteins , Cyclins/genetics , Gene Expression Regulation , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins c-myc/physiology , Repressor Proteins , Acetylation , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Cyclin D2 , DNA-Binding Proteins/physiology , HL-60 Cells , Histones/metabolism , Humans , RNA, Messenger/biosynthesis , RNA-Binding Proteins/physiology , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...