Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
BMC Dev Biol ; 16(1): 30, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27582005

ABSTRACT

BACKGROUND: Wnt11 is a member of the Wnt family of secreted signals controlling the early steps in ureteric bud (UB) branching. Due to the reported lethality of Wnt11 knockout embryos in utero, its role in later mammalian kidney organogenesis remains open. The presence of Wnt11 in the emerging tubular system suggests that it may have certain roles later in the development of the epithelial ductal system. RESULTS: The Wnt11 knockout allele was backcrossed with the C57Bl6 strain for several generations to address possible differences in penetrance of the kidney phenotypes. Strikingly, around one third of the null mice with this inbred background survived to the postnatal stages. Many of them also reached adulthood, but urine and plasma analyses pointed out to compromised kidney function. Consistent with these data the tubules of the C57Bl6 Wnt11 (-/-) mice appeared to be enlarged, and the optical projection tomography indicated changes in tubular convolution. Moreover, the C57Bl6 Wnt11 (-/-) mice developed secondary glomerular cysts not observed in the controls. The failure of Wnt11 signaling reduced the expression of several genes implicated in kidney development, such as Wnt9b, Six2, Foxd1 and Hox10. Also Dvl2, an important PCP pathway component, was downregulated by more than 90 % due to Wnt11 deficiency in both the E16.5 and NB kidneys. Since all these genes take part in the control of UB, nephron and stromal progenitor cell differentiation, their disrupted expression may contribute to the observed anomalies in the kidney tubular system caused by Wnt11 deficiency. CONCLUSIONS: The Wnt11 signal has roles at the later stages of kidney development, namely in coordinating the development of the tubular system. The C57Bl6 Wnt11 (-/-) mouse generated here provides a model for studying the mechanisms behind tubular anomalies and glomerular cyst formation.


Subject(s)
Kidney Glomerulus/abnormalities , Kidney Tubules/abnormalities , Wnt Proteins/genetics , Wnt Proteins/metabolism , Animals , Cell Differentiation , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Kidney Glomerulus/embryology , Kidney Tubules/embryology , Mice , Mice, Knockout , Signal Transduction
2.
Mol Carcinog ; 55(2): 170-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25641046

ABSTRACT

The epidermal growth factor receptor (EGFR) is involved in the regulation of various cellular processes and dysregulation of its signalling plays a critical role in the etiology of a variety of malignancies like breast cancer. At the same time, elevated levels of urokinase (uPA), its receptor uPAR, and other components of the plasminogen activation system are found to be correlated with a poor prognosis in breast cancer. Interestingly, EGFR appears to participate in transducing the signal generated upon binding of uPA to uPAR. However, whether uPA signalling would thereby interfere with ligand-driven EGFR signalling was not described before. Therefore, it was the aim of the present study to investigate the combined effects of uPA and EGF in the low invasive and high invasive breast adenocarcinoma cell lines MCF-7 and MDA-MB-231, respectively. Simultaneous exposure of cells to both signals negatively affected ERK1/2 and AKT activation whereas positive effects on p38 and Src kinase phosphorylation were noted in both cell lines. Furthermore, uPA attenuated the mitogenic effect of EGF on cellular proliferation, invasion and motility in both MCF-7 and MDA-MB-231 cells. Experiments with the uPA amino terminal fragment (ATF) revealed that the negative effects of uPA were independent from its protease activity. Together, these data suggest that enhanced levels of uPA in breast cancer modulate the mitogenic effects of EGF and thus, this knowledge may help to better understand breast cancer pathogenesis as well as to develop new therapeutic options.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , EGF Family of Proteins/pharmacology , Urokinase-Type Plasminogen Activator/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells
3.
Exp Cell Res ; 332(2): 163-78, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25645944

ABSTRACT

The indifferent mammalian embryonic gonad generates an ovary or testis, but the factors involved are still poorly known. The Wnt-4 signal represents one critical female determinant, since its absence leads to partial female-to-male sex reversal in mouse, but its signalling is as well implicated in the testis development. We used the Wnt-4 deficient mouse as a model to identify candidate gonadogenesis genes, and found that the Notum, Phlda2, Runx-1 and Msx1 genes are typical of the wild-type ovary and the Osr2, Dach2, Pitx2 and Tacr3 genes of the testis. Strikingly, the expression of these latter genes becomes reversed in the Wnt-4 knock-out ovary, suggesting a role in ovarian development. We identified the transcription factor Runx-1 as a Wnt-4 signalling target gene, since it is expressed in the ovary and is reduced upon Wnt-4 knock-out. Consistent with this, introduction of the Wnt-4 signal into early ovary cells ex vivo induces Runx-1 expression, while conversely Wnt-4 expression is down-regulated in the absence of Runx-1. We conclude that the Runx-1 gene can be a Wnt-4 signalling target, and that Runx-1 and Wnt-4 are mutually interdependent in their expression. The changes in gene expression due to the absence of Wnt-4 in gonads reflect the sexually dimorphic role of this signal and its complex gene network in mammalian gonad development.


Subject(s)
Gene Expression Regulation, Developmental , Ovary/metabolism , Wnt4 Protein/physiology , Animals , Base Sequence , Binding Sites , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Gene Expression , Male , Mice, Knockout , Ovary/embryology , Sex Determination Processes/genetics , Tissue Culture Techniques , Wnt Signaling Pathway
4.
Cell Signal ; 20(1): 154-62, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18006272

ABSTRACT

Thrombopoietin (TPO), a hematopoietic growth factor regulating platelet production, and its receptor (TPOR) were recently shown to be expressed in the brain where they exert proapoptotic activity. Here we used PC12 cells, an established model of neuronal differentiation, to investigate the effects of TPO on neuronal survival and differentiation. These cells expressed TPOR mRNA. TPO increased cell death in neuronally differentiated PC12 cells but had no effect in undifferentiated cells. Surprisingly, TPO inhibited nerve growth factor (NGF)-induced differentiation of PC12 cells in a dose- and time-dependent manner. This inhibition was dependent on the activity of Janus kinase-2 (JAK2). Using phospho-kinase arrays and Western blot we found downregulation of the NGF-stimulated phosphorylation of the extracellular signal-regulated kinase p42ERK by TPO with no effect on phosphorylation of Akt or stress kinases. NGF-induced phosphorylation of ERK-activating kinases, MEK1/2 and C-RAF was also reduced by TPO while NGF-induced RAS activation was not attenuated by TPO treatment. In contrast to its inhibitory effects on NGF signalling, TPO had no effect on epidermal growth factor (EGF)-stimulated ERK phosphorylation or proliferation of PC12 cells. Our data indicate that TPO via activation of its receptor-bound JAK2 delays the NGF-dependent acquisition of neuronal phenotype and decreases neuronal survival by suppressing NGF-induced ERK activity.


Subject(s)
Cell Differentiation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Nerve Growth Factor/physiology , Neurons/cytology , Signal Transduction , Thrombopoietin/physiology , Animals , Cell Survival/drug effects , Epidermal Growth Factor/physiology , MAP Kinase Kinase 1/metabolism , Neurons/physiology , PC12 Cells , Proto-Oncogene Proteins c-raf/metabolism , Rats , ras Proteins/metabolism
5.
Cancer Res ; 79(16): 4042-4056, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31142511

ABSTRACT

The EGFR adaptor protein, CIN85, has been shown to promote breast cancer malignancy and hypoxia-inducible factor (HIF) stability. However, the mechanisms underlying cancer promotion remain ill defined. Here we show that CIN85 is a novel binding partner of the main HIF-prolyl hydroxylase, PHD2, but not of PHD1 or PHD3. Mechanistically, the N-terminal SRC homology 3 domains of CIN85 interacted with the proline-arginine-rich region within the N-terminus of PHD2, thereby inhibiting PHD2 activity and HIF degradation. This activity is essential in vivo, as specific loss of the CIN85-PHD2 interaction in CRISPR/Cas9-edited cells affected growth and migration properties, as well as tumor growth in mice. Overall, we discovered a previously unrecognized tumor growth checkpoint that is regulated by CIN85-PHD2 and uncovered an essential survival function in tumor cells by linking growth factor adaptors with hypoxia signaling. SIGNIFICANCE: This study provides unprecedented evidence for an oxygen-independent mechanism of PHD2 regulation that has important implications in cancer cell survival. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/16/4042/F1.large.jpg.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Triple Negative Breast Neoplasms/pathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , Cell Line, Tumor , Female , HEK293 Cells , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Mice, Nude , Protein Interaction Domains and Motifs , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
6.
Dis Model Mech ; 10(12): 1503-1515, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29084770

ABSTRACT

Three-dimensional (3D) organoids provide a new way to model various diseases, including cancer. We made use of recently developed kidney-organ-primordia tissue-engineering technologies to create novel renal organoids for cancer gene discovery. We then tested whether our novel assays can be used to examine kidney cancer development. First, we identified the transcriptomic profiles of quiescent embryonic mouse metanephric mesenchyme (MM) and of MM in which the nephrogenesis program had been induced ex vivo The transcriptome profiles were then compared to the profiles of tumor biopsies from renal cell carcinoma (RCC) patients, and control samples from the same kidneys. Certain signature genes were identified that correlated in the developmentally induced MM and RCC, including components of the caveolar-mediated endocytosis signaling pathway. An efficient siRNA-mediated knockdown (KD) of Bnip3, Gsn, Lgals3, Pax8, Cav1, Egfr or Itgb2 gene expression was achieved in mouse RCC (Renca) cells. The live-cell imaging analysis revealed inhibition of cell migration and cell viability in the gene-KD Renca cells in comparison to Renca controls. Upon siRNA treatment, the transwell invasion capacity of Renca cells was also inhibited. Finally, we mixed E11.5 MM with yellow fluorescent protein (YFP)-expressing Renca cells to establish chimera organoids. Strikingly, we found that the Bnip3-, Cav1- and Gsn-KD Renca-YFP+ cells as a chimera with the MM in 3D organoid rescued, in part, the RCC-mediated inhibition of the nephrogenesis program during epithelial tubules formation. Altogether, our research indicates that comparing renal ontogenesis control genes to the genes involved in kidney cancer may provide new growth-associated gene screens and that 3D RCC-MM chimera organoids can serve as a novel model with which to investigate the behavioral roles of cancer cells within the context of emergent complex tissue structures.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Renal Cell/pathology , Chimera/metabolism , Genetic Association Studies , Kidney Neoplasms/pathology , Kidney/pathology , Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Cell Differentiation , Cell Line, Tumor , Cell Movement/genetics , Coculture Techniques , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , HEK293 Cells , Humans , Kidney Neoplasms/genetics , Mice , Neoplasm Invasiveness , Nephrons/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Transfection , Tumor Stem Cell Assay
7.
Cancers (Basel) ; 8(6)2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27322325

ABSTRACT

Renal cell carcinoma (RCC) accounts for 90% of all kidney cancers. Due to poor diagnosis, high resistance to the systemic therapies and the fact that most RCC cases occur sporadically, current research switched its focus on studying the molecular mechanisms underlying RCC. The aim is the discovery of new effective and less toxic anti-cancer drugs and novel diagnostic markers. Besides the PI3K/Akt/mTOR, HGF/Met and VHL/hypoxia cellular signaling pathways, the involvement of the Wnt/ß-catenin pathway in RCC is commonly studied. Wnt signaling and its targeted genes are known to actively participate in different biological processes during embryonic development and renal cancer. Recently, studies have shown that targeting this pathway by alternating/inhibiting its intracellular signal transduction can reduce cancer cells viability and inhibit their growth. The targets and drugs identified show promising potential to serve as novel RCC therapeutics and prognostic markers. This review aims to summarize the current status quo regarding recent research on RCC focusing on the involvement of the Wnt/ß-catenin pathway and how its understanding could facilitate the identification of potential therapeutic targets, new drugs and diagnostic biomarkers.

8.
Thromb Haemost ; 113(5): 988-98, 2015 May.
Article in English | MEDLINE | ID: mdl-25694133

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is the major and most specific acting urokinase (uPA) and tissue plasminogen activator (tPA) inhibitor. Apart from its function in the fibrinolytic system, PAI-1 was also found to contribute to processes like tissue remodelling, angiogenesis, and tumour progression. However, the role of PAI-1 in those processes remains largely controversial with respect to the influence of PAI-1 on cell signalling pathways. Although PAI-1 does not possess its own cellular receptor, it can be bound to low-density lipoprotein receptor-related protein 1 (LRP1) which was proposed to modulate the ß-catenin pathway. Therefore, we used wild-type mouse embryonic fibroblasts (MEFs), and MEFs deficient of LRP1 to study PAI-1 as modulator of the ß-catenin pathway. We found that PAI-1 influences MEF proliferation and motility in a LRP1-dependent manner and that ß-catenin is important for that response. In addition, expression of ß-catenin and ß-catenin-dependent transcriptional activity were induced by PAI-1 in wild type MEFs, but not in LRP1-deficient cells. Moreover, PAI-1-induced ERK1/2 activation was more prominent in the LRP1-deficient cells and interestingly knockdown of ß-catenin abolished this effect. Together, the data of the current study show that PAI-1 can promote cell migration via LRP1-dependent activation of the ß-catenin and ERK1/2 MAPK pathway which may be important in stage-specific treatment of human diseases associated with high PAI-1 levels.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, LDL/metabolism , Serpin E2/metabolism , Tissue Plasminogen Activator/metabolism , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Movement , Cell Proliferation , Fibroblasts/metabolism , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , Transcription, Genetic , Urokinase-Type Plasminogen Activator/metabolism
9.
Antioxid Redox Signal ; 6(4): 777-91, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15242559

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is the major physiological inhibitor of urokinase-type and tissue-type plasminogen activators. It has gained special interest among clinicians because a number of pathological conditions, such as myocardial infarction, atherosclerosis, thrombosis, several types of cancer, and the metabolic syndrome, as well as type 2 diabetes mellitus, are associated with increased PAI-1 levels. Interestingly, a number of these diseases are also accompanied by oxidative stress and the enhanced production of reactive oxygen species or tissue hypoxia. This article tries to summarize some aspects leading to enhanced PAI-1 production under oxidative stress or hypoxia.


Subject(s)
Hypoxia , Oxidative Stress , Plasminogen Activator Inhibitor 1/metabolism , Animals , Antioxidants/metabolism , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinases/metabolism , Plasminogen Activator Inhibitor 1/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/toxicity , Signal Transduction/physiology , Transcription Factors/metabolism
10.
Antioxid Redox Signal ; 6(5): 802-10, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15345139

ABSTRACT

Heme oxygenase (HO)-1 is the inducible isoform of the rate-limiting enzyme of heme degradation, which is up-regulated by a host of stress stimuli. The bacterial cell membrane component lipopolysaccharide (LPS) is a prototypical activator of monocytic cells. Here, it is shown that LPS induced the endogenous HO-1 gene expression in RAW264.7 monocytic cells. To investigate the molecular mechanisms of HO-1 gene induction by LPS, we performed transfection experiments with reporter gene constructs containing sequences of the proximal rat HO-1 gene promoter. Deletion and mutation analysis indicated that a cyclic AMP response element/activator protein-1 site (-664/-657), but not an E-box motif (-47/-42), played a major role for LPS-dependent HO-1 gene induction. Up-regulation of HO-1 promoter activity by LPS was decreased by pharmacological nuclear factor-kappaB (NF-kappaB) inhibitors and by cotransfected expression vectors with dominant negative isoforms of NF-kappaB-inducing kinase, inhibitor of NF-kappaB (IkappaB) kinase beta, and IkappaBalpha. Moreover, the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 and overexpressed dominant negative p38beta decreased, whereas dominant negative p38delta increased, LPS-dependent induction of HO-1 gene expression. The results suggest that the NF-kappaB and p38 MAPK signaling pathways mediate the LPS-dependent induction of HO-1 gene expression via DNA sequences of the proximal promoter region.


Subject(s)
Enzyme Induction , Heme Oxygenase (Decyclizing)/metabolism , Lipopolysaccharides/metabolism , MAP Kinase Signaling System , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Enzyme Activation , Genes, Reporter , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Membrane Proteins , Mice , Monocytes/cytology , Monocytes/metabolism , NF-kappa B/antagonists & inhibitors , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , NF-kappaB-Inducing Kinase
11.
Antioxid Redox Signal ; 19(17): 2157-96, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-23458328

ABSTRACT

Reactive oxygen species (ROS) exert various biological effects and contribute to signaling events during physiological and pathological processes. Enhanced levels of ROS are highly associated with different tumors, a Western lifestyle, and a nutritional regime. The supplementation of food with traditional antioxidants was shown to be protective against cancer in a number of studies both in vitro and in vivo. However, recent large-scale human trials in well-nourished populations did not confirm the beneficial role of antioxidants in cancer, whereas there is a well-established connection between longevity of several human populations and increased amount of antioxidants in their diets. Although our knowledge about ROS generators, ROS scavengers, and ROS signaling has improved, the knowledge about the direct link between nutrition, ROS levels, and cancer is limited. These limitations are partly due to lack of standardized reliable ROS measurement methods, easily usable biomarkers, knowledge of ROS action in cellular compartments, and individual genetic predispositions. The current review summarizes ROS formation due to nutrition with respect to macronutrients and antioxidant micronutrients in the context of cancer and discusses signaling mechanisms, used biomarkers, and its limitations along with large-scale human trials.


Subject(s)
Antioxidants/physiology , Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Vitamins/physiology , Animals , Antineoplastic Agents/administration & dosage , Antioxidants/administration & dosage , Carcinogenesis/metabolism , Diet , Dietary Supplements , Humans , Neoplasms/drug therapy , Oxidative Stress , Vitamins/administration & dosage
12.
Antioxid Redox Signal ; 13(4): 413-24, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20136501

ABSTRACT

The plasminogen activator inhibitor-1 (PAI-1) expression can be enhanced by hypoxia and various stimuli associated with oxidative stress. Among the FOXO transcription factors, FOXO4 appears to be crucial in the response against oxidative stress. Therefore, it was the aim of this study to investigate the role of peroxide-induced oxidative stress and FOXO4 on PAI-1 expression under normoxia and hypoxia. Treatment of cells with hydrogen peroxide increased PAI-1 mRNA, protein, and promoter activity, and knocking down FOXO4 abolished the peroxide-dependent PAI-1 induction. PAI-1 promoter reporter gene assays revealed that the peroxide and FOXO4-dependent induction was mediated through the HIF-1 and CREB-binding HRE within the PAI-1 promoter. Western blot analyses then indicated that peroxide and FOXO4 downregulated HIF-1alpha levels, whereas CREB levels were increased. Chromatin immunoprecipitations showed that FOXO4 did not bind the PAI-1 promoter, whereas CREB binding was enhanced on FOXO4 overexpression. In addition, knockdown of CREB abolished the FOXO4-mediated PAI-1 induction. Together, these findings provide the first evidence that oxidative stress and FOXO4 induce PAI-1 expression through an indirect mechanism involving modulation of HIF-1alpha and CREB protein levels and that enhanced CREB binding to the PAI-1 promoter is critical for the PAI-1 induction under oxidative stress.


Subject(s)
CREB-Binding Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Transcription Factors/metabolism , Binding Sites , Biological Transport/genetics , Biological Transport/physiology , Blotting, Northern , Blotting, Western , CREB-Binding Protein/genetics , Cell Cycle Proteins , Cells, Cultured , Chromatin Immunoprecipitation , Forkhead Transcription Factors , Hep G2 Cells , Humans , Hydrogen Peroxide/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Binding/physiology , RNA, Small Interfering , Transcription Factors/genetics
13.
Thromb Haemost ; 103(5): 901-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20216986

ABSTRACT

Increased levels of plasminogen activator inhibitor-1 (PAI-1) indicate an enhanced risk of ischaemic/hypoxic cardiovascular events and a poor prognosis. The expression of PAI-1 can be induced by various stimuli including hypoxia, insulin and insulin-like growth factor 1 (IGF-1). The hypoxia-inducible factor-1 (HIF-1) is critical for hypoxia or insulin/IGF-1 mediated PAI-1 induction, but the components involved in merging the signals are not known so far. The adaptor/scaffold protein Ruk/CIN85 may be a candidate since it plays important roles in the regulation of processes associated with cardiovascular and oncological diseases such as downregulation of receptor tyrosine kinases, apoptosis, adhesion and invasion. Therefore, it was the aim of this study to investigate the involvement of Ruk/CIN85 in the regulation of PAI-1 expression. It was found that Ruk/CIN85 induced PAI-1 mRNA and protein expression both under normoxia and hypoxia. The induction of PAI-1 expression by Ruk/CIN85 occurred at the transcriptional level since the half-life of PAI-1 mRNA was not affected in cells overexpressing Ruk/CIN85 and reporter gene assays using wild-type and mutant human PAI-1 promoter luciferase constructs showed that the hypoxia responsive element was responsible for Ruk/CIN85 effects. Further, knocking down HIF-1alpha abolished not only the hypoxia-dependent but also the Ruk/CIN85-dependent PAI-1 induction. In addition, transient or stable overexpression of Ruk/CIN85 also induced HIF-1alpha protein levels and HIF-1 activity and knocking down Ruk/CIN85 reversed these effects. Thereby, Ruk/CIN85 interfered with the proline hydroxylation-dependent HIF-1alpha protein destabilisation. Together, these results provide the first evidence that Ruk/CIN85 induces PAI-1 expression via modulation of HIF-1alpha stability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Cloning, Molecular , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Plasminogen Activator Inhibitor 1/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Transcriptional Activation/genetics , Transgenes/genetics
14.
Antioxid Redox Signal ; 10(7): 1163-74, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18331200

ABSTRACT

Heme oxygenase-1 is the rate-limiting enzyme for the degradation of the prooxidant heme. Previously, we showed that an E-box within the HO-1 promoter is crucial for the regulation of HO-1 expression in primary hepatocytes. Further to investigate the importance of this E-box, we determined the regulatory capacity of the E-box-binding factor USF-2 in primary cells in comparison with transformed cell lines. We found that HO-1 expression was inhibited by USF-2 in primary cells, whereas it was induced in tumor cell lines. Mutation of either the E-box or the AP-1 site within the HO-1 promoter only partially affected the USF-dependent regulation. However, this regulation was dramatically reduced in tumor cells and completely abolished in primary cells transfected with an HO-1 promoter construct containing mutations in both the E-box and the AP-1 site, suggesting that AP-1 factors and USF-2 may act in a cooperative manner. Indeed, protein-protein interaction studies revealed that USF proteins interacted with Fra-1. Further, the USF-dependent HO-1 promoter activity was not detectable with an USF-2 mutant lacking residues of the USF-specific region (USR) or the transactivation domain encoded by exon 4. Together, these data suggest that USF-2 has opposite regulatory roles for HO-1 gene expression in primary cells and tumor cell lines.


Subject(s)
Heme Oxygenase-1/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Upstream Stimulatory Factors/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Electrophoretic Mobility Shift Assay , HeLa Cells , Heme Oxygenase-1/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Immunoprecipitation , Male , Models, Genetic , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Wistar , Upstream Stimulatory Factors/genetics
15.
Neurochem Res ; 31(10): 1219-30, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17021950

ABSTRACT

Correct timing and spatial location of growth factor expression is critical for undisturbed brain development and functioning. In terminally differentiated cells distinct biological responses to growth factors may depend on cell type specific activation of signalling cascades. We show that the hematopoietic growth factors thrombopoietin (TPO) and granulocyte colony-stimulating factor (GCSF) exert cell type specific effects on survival, proliferation and the degree of phosphorylation of Akt1, ERK1/2 and STAT3 in rat hippocampal neurons and cortical astrocytes. In neurons, TPO induced cell death and selectively activated ERK1/2. GCSF protected neurons from TPO- and hypoxia-induced cell death via selective activation of Akt1. In astrocytes, neither TPO nor GCSF had any effect on cell viability but inhibited proliferation. This effect was accompanied by activation of ERK1/2 and inhibition of STAT3 activity. A balance between growth factors, their receptors and signalling proteins may play an important role in regulation of neural cell survival.


Subject(s)
Astrocytes/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Thrombopoietin/pharmacology , Animals , Astrocytes/metabolism , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , STAT3 Transcription Factor/metabolism
16.
J Hepatol ; 45(1): 72-80, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16510205

ABSTRACT

BACKGROUND/AIMS: Heme oxygenase-1 (HO-1) can be induced by various stimuli, one of which is interleukin-6 (IL-6). Therefore, the aim of this study was to elucidate the molecular mechanisms responsible for IL-6-dependent HO-1 induction in the liver. METHODS: The IL-6-dependent HO-1 regulation in rat primary hepatocytes and HepG2 hepatoma cells was studied by Northern and Western blot analyses, HO-1 promoter reporter gene assays and EMSA. RESULTS: The HO-1 expression was transcriptionally induced by IL-6 in a time- and dose-dependent manner. Activation of signal transducers and activators of transcription (STAT) factors by the IL-6 receptor was crucial for HO-1 induction. By contrast, negative regulation of HO-1 expression appeared to be mediated through the SH2-domain-containing tyrosine phosphatase-2 (SHP2)/ suppressors of cytokine signaling-3 (SOCS3) binding site within the gp130 IL-6 receptor subunit. Among the three putative STAT binding elements (SBE) in the HO-1 promoter, only the distal one was functional and when deleted, the remaining Luc induction was completely obliterated by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. CONCLUSIONS: The HO-1 SBE3 mediates HO-1 gene induction by IL-6 mainly via activation of the Jak/STAT pathway.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase (Decyclizing)/genetics , Hepatocytes/enzymology , Interleukin-6/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Northern , Cell Culture Techniques , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Kinetics , Male , Plasmids , RNA, Messenger/genetics , Rats , Rats, Wistar , Signal Transduction/immunology , Transcriptional Activation
17.
Lab Invest ; 85(3): 376-87, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15640832

ABSTRACT

Heme oxygenase-1 (HO-1) is the inducible isoform of an enzyme family responsible for heme degradation and was suggested to be involved in the acute phase response in the liver. However, the mechanisms of the HO-1 regulation under inflammatory conditions are poorly understood. Therefore, the purpose of the current work was to study the expression of HO-1 in the liver and other organs of rats with a localized inflammation after intramuscular injection of turpentine oil (TO). Since interleukin-6 (IL-6) is known to be a principal mediator of inflammation, the levels of this cytokine were also estimated in the animal model used. HO-1 and IL-6 expression was evaluated by Northern blot, in situ hybridization, Western blot, immunohistochemistry and enzyme-linked immunosorbent assay. In the liver and injured muscle, the HO-1 mRNA levels were dramatically increased 4-6 h after TO administration. HO-1 protein levels in the liver were elevated starting from 6-12 h after the treatment. In other internal organs such as the heart, kidney and large intestine, only a slight induction of HO-1 mRNA was observed. IL-6-specific transcripts appeared only in the injured muscle and were in accordance with serum levels of IL-6. In turn, temporal expression of IL-6 in the muscle and circulatory IL-6 levels correlated well with HO-1 expression in the liver and injured muscle. In the liver of control rats HO-1 protein was detected in Kupffer cells, while in TO-injected rats also hepatocytes became strongly HO-1 positive. Conversely, in the injured muscle, HO-1 immunoreactivity was attributed only to macrophages. Our data demonstrate that during localized inflammation HO-1 expression was rapidly and strongly induced in macrophages of injured muscle and in hepatocytes, and IL-6 derived from injured muscle seems to be responsible for the HO-1 induction in the liver.


Subject(s)
Acute-Phase Reaction/enzymology , Chemical and Drug Induced Liver Injury/enzymology , Heme Oxygenase (Decyclizing)/biosynthesis , Hepatocytes/enzymology , Interleukin-6/metabolism , Kupffer Cells/enzymology , Turpentine/toxicity , Acute-Phase Reaction/immunology , Acute-Phase Reaction/pathology , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/immunology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Enzymologic , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Hepatocytes/drug effects , Hepatocytes/immunology , Injections, Intramuscular , Interleukin-6/immunology , Kupffer Cells/drug effects , Kupffer Cells/immunology , Male , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , RNA, Messenger/genetics , Rats , Rats, Wistar , Turpentine/administration & dosage , Up-Regulation
18.
J Biol Chem ; 278(20): 17927-36, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12637567

ABSTRACT

Heme oxygenase-1 (HO-1) gene expression is induced by various oxidative stress stimuli including sodium arsenite. Since mitogen-activated protein kinases (MAPKs) are involved in stress signaling we investigated the role of arsenite and MAPKs for HO-1 gene regulation in primary rat hepatocytes. The Jun N-terminal kinase (JNK) inhibitor SP600125 decreased sodium arsenite-mediated induction of HO-1 mRNA expression. HO-1 protein and luciferase activity of reporter gene constructs with -754 bp of the HO-1 promoter were induced by overexpression of kinases of the JNK pathway and MKK3. By contrast, overexpression of Raf-1 and ERK2 did not affect expression whereas overexpression of p38alpha, beta, and delta decreased and p38gamma increased HO-1 expression. Electrophoretic mobility shift assays (EMSA) revealed that a CRE/AP-1 element (-668/-654) bound c-Jun, a target of the JNK pathway. Deletion or mutation of the CRE/AP-1 obliterated the JNK- and c-Jun-dependent up-regulation of luciferase activity. EMSA also showed that an E-box (-47/-42) was bound by a putative p38 target c-Max. Mutation of the E-box strongly reduced MKK3, p38 isoform-, and c-Max-dependent effects on luciferase activity. Thus, the HO-1 CRE/AP-1 element mediates HO-1 gene induction via activation of JNK/c-Jun whereas p38 isoforms act through a different mechanism via the E-box.


Subject(s)
Gene Expression Regulation, Enzymologic , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase (Decyclizing)/genetics , Hepatocytes/enzymology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Transcription, Genetic , Animals , Anthracenes/pharmacology , Arsenites/pharmacology , Base Sequence , Cell Nucleus/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1 , Luciferases/metabolism , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase 1/metabolism , Models, Genetic , Molecular Sequence Data , Mutation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sodium Compounds/pharmacology , Transcriptional Activation , Transfection , Up-Regulation , p38 Mitogen-Activated Protein Kinases
19.
Blood ; 101(3): 907-14, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12393531

ABSTRACT

The expression of the plasminogen activator inhibitor-1 (PAI-1) gene is enhanced by insulin both in vivo and in various cell types. Because insulin exerts a number of its biologic activities via the phosphatidylinositol 3-kinase and protein kinase B (PI3K/PKB) signaling pathway, it was the aim of the present study to investigate the role of the PI3K/PKB pathway in the expression of the PAI-1 gene and to identify the insulin responsive promoter sequences. It was shown that the induction of PAI-1 mRNA and protein expression by insulin and mild hypoxia could be repressed by the PI3K inhibitor wortmannin. Overexpression of a constitutively active PKB led to induction of PAI-1 mRNA expression and of luciferase (Luc) activity from a gene construct containing 766 bp of the rat PAI-1 promoter. Mutation of the hypoxia response elements (HRE-1 and HRE-2) in rat PAI-1 promoter, which could bind hypoxia inducible factor-1 (HIF-1), abolished the induction of PAI-1 by insulin and PKB. Insulin and the constitutive active PKB also induced Luc expression in cells transfected with the pGl3EPO-HRE Luc construct, containing 3 copies of the HRE from the erythropoietin gene in front of the SV40 promoter. Furthermore, insulin and the active PKB enhanced all 3 HIF alpha-subunit protein levels and HIF-1 DNA-binding activity, as shown by electrophoretic mobility shift assays (EMSAs). Thus, the insulin-dependent activation of the PAI-1 gene expression can be mediated via the PI3K/PKB pathway and the transcription factor HIF-1 binding to the HREs in the PAI-1 gene promoter.


Subject(s)
DNA-Binding Proteins/genetics , Hypoxia/genetics , Insulin/pharmacology , Nuclear Proteins/genetics , Plasminogen Activator Inhibitor 1/biosynthesis , Protein Serine-Threonine Kinases , Response Elements , Animals , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Male , Phosphatidylinositol 3-Kinases/metabolism , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Rats, Wistar , Transcription Factors/drug effects , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL