Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
2.
Biochem Pharmacol ; 158: 1-12, 2018 12.
Article in English | MEDLINE | ID: mdl-30248327

ABSTRACT

In its classical genomic mode of action, the aryl hydrocarbon receptor (AhR) acts as a ligand activated transcription factor regulating expression of target genes such as CYP1A1 and CYP1B1. Some ligands may also trigger more rapid nongenomic responses through AhR, including calcium signaling (Ca2+). In the present study we observed that pyrene induced a relatively rapid increase in intracellular Ca2+-concentrations ([Ca2+]i) in human microvascular endothelial cells (HMEC-1) and human embryonic kidney cells (HEK293) that was attenuated by AhR-inhibitor treatment and/or transient AhR knockdown by RNAi. In silico molecular docking based on homology models, suggested that pyrene is not able to bind to the human AhR in the agonist conformation. Instead, pyrene docked in the antagonist conformation of the AhR PAS-B binding pocket, although the interaction differed from antagonists such as GNF-351 and CH223191. Accordingly, pyrene did not induce CYP1A1 or CYP1B1, but suppressed CYP1-expression by benzo[a]pyrene (B[a]P) in HMEC-1 cells, confirming that pyrene act as an antagonist of AhR-induced gene expression. Use of pharmacological inhibitors and Ca2+-free medium indicated that the pyrene-induced AhR nongenomic [Ca2+]i increase was initiated by Ca2+-release from intracellular stores followed by a later phase of extracellular Ca2+-influx, consistent with store operated calcium entry (SOCE). These effects was accompanied by an AhR-dependent reduction in ordered membrane lipid domains, as determined by di-4-ANEPPDHQ staining. Addition of cholesterol inhibited both the pyrene-induced [Ca2+]i-increase and alterations in membrane lipid order. In conclusion, we propose that pyrene binds to AhR, act as an antagonist of the canonical genomic AhR/Arnt/CYP1-pathway, reduces ordered membrane lipid domains, and activates AhR nongenomic Ca2+-signaling from intracellular stores.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium Signaling/physiology , Pyrenes/metabolism , Pyrenes/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Azo Compounds/chemistry , Azo Compounds/metabolism , Azo Compounds/pharmacology , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/chemistry , Binding Sites , Calcium Signaling/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HEK293 Cells , Humans , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Molecular Docking Simulation/methods , Protein Structure, Secondary , Purines/chemistry , Purines/metabolism , Purines/pharmacology , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrenes/chemistry , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/chemistry
3.
Inflamm Bowel Dis ; 24(3): 593-600, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29462394

ABSTRACT

Background: The colon and rectum are continuously exposed to oxidative stress that generates reactive oxygen species, which are a major cause of DNA double-strand breaks (DSB). Furthermore, chronic inflammatory diseases such as ulcerative colitis (UC) are characterized by an excess of reactive nitrogen species that can also lead to DNA double-strand breakage and genomic instability. We investigated the expression of the nuclear casein kinase and cyclin-dependent kinase substrate 1 (NUCKS1) protein in UC and sporadic colorectal cancer (CRC) due to its involvement in both DNA double-strand break repair and inflammatory signaling. Methods: NUCKS1 expression and expression of the DNA double-strand break marker gamma-H2AX (γH2AX) were assessed in formalin-fixed, paraffin-embedded UC and CRC patient biopsies using peroxidase immunohistochemistry. Expression levels for both proteins were evaluated together with previously published expression-level data for hTERT and TP53 proteins in the same material. Results: Nondysplastic UC lesions had 10-fold lower γH2AX expression and approximately 4-fold higher NUCKS1 expression compared with sporadic CRC, indicating minimal DNA DSB damage and heightened DNA DSB repair in these lesions, respectively. NUCKS1 expression in UC tended to decrease with increasing grades of dysplasia, whereas γH2AX, hTERT, and TP53 expression tended to increase with increasing grades of dysplasia. The highest γH2AX expression was seen in sporadic CRC, indicating considerable DNA DSB damage, whereas the highest NUCKS1 expression and hTERT expression were seen in nondysplastic UC. Conclusions: Overall, our data suggest that NUCKS1 may be involved in DNA DSB repair and/or inflammatory signaling in UC, but a more thorough investigation of both pathways in UC is warranted.


Subject(s)
Colitis, Ulcerative/metabolism , Colorectal Neoplasms/metabolism , DNA Breaks, Double-Stranded , Histones/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Aged , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Genetic Markers , Genomic Instability , Histones/genetics , Humans , Immunohistochemistry , Male , Nuclear Proteins/genetics , Phosphoproteins/genetics , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Scand J Gastroenterol ; 49(1): 99-108, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24188385

ABSTRACT

OBJECTIVE: Liver regeneration following hepatectomy can stimulate the growth of hepatocellular carcinoma (HCC), and major hepatectomy can be associated with activation of hepatic progenitor cells (HPCs). The aim of this study was to evaluate how HPCs influence the malignant potential of tumor cells in vitro and HCC tumor growth after surgery in a rodent model. MATERIAL AND METHODS: Hepatoma cells (JM1) were cultured with conditioned medium (CM) from syngeneic HPCs (WB-F344). Growth rate, resistance to Adriamycin, and expression patterns for invasiveness and stemness were compared with naïve JM1. Microscopic HCC tumors from naïve JM1 or JM1 cultured with CM were inoculated in Fischer 344 rats undergoing 70% hepatectomy with or without simultaneous infusion of WB-F344. Tumor growth and invasiveness-related factors were compared. Buffalo rats were induced with Morris hepatoma cells. Liver tissue from both in vivo models was examined with regard to activation of cells with progenitor-like phenotype. RESULTS: Co-culture with CM resulted in an increased resistance to Adriamycin and enhanced expressions of α-FP, MMP9, ABCG2, CD133, and SOX2, as well as the activation of ERK, AKT, WNT, and TGF-ß1 pathways. Tumor size and metastases were significantly higher in groups with co-cultured cells or HPCs infusion. After 70% hepatectomy and tumor implantation, cells positive for α-FP, CK19, and CD133 were found, thus suggesting a progenitor-like phenotype in the setting of epithelial-mesenchymal transition. CONCLUSION: HPCs have a marked effect on HCC cells in vitro and appear to stimulate the growth and malignant potential of experimental HCC tumors.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Regeneration , Stem Cells/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Cytokine Receptor Common beta Subunit/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Gene Expression , Hepatectomy , Humans , Liver/physiology , Liver/surgery , Liver Neoplasms, Experimental/pathology , MAP Kinase Signaling System , Matrix Metalloproteinase 9/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred BUF , Rats, Inbred F344 , SOXB1 Transcription Factors/genetics , Transforming Growth Factor beta1/metabolism , Tumor Burden , Wnt Signaling Pathway , alpha-Fetoproteins/metabolism
7.
Cell Physiol Biochem ; 32(3): 511-22, 2013.
Article in English | MEDLINE | ID: mdl-24008581

ABSTRACT

BACKGROUND/AIMS: EGF receptor is a main participant in the regulation of liver regeneration. In primary hepatocyte cultures, EGF or TGFα binding to EGF receptor activates Erk1/2 and PI3K pathways, induces cyclin D1 and thus initiates DNA synthesis. We have explored mechanisms by which prolonged EGF receptor activation induces hepatocyte proliferation. METHODS: EGF receptor activation, as well as Erk1/2 and PI3K signaling were explored in EGF-stimulated primary hepatocyte cultures by Western blotting and immunocytochemistry. TGFα release to the medium was quantified by ELISA. Effects of a neutralizing antibody to TGFα on EGF receptor signaling and proliferation were explored. RESULTS: Inhibitors of PI3K or Erk1/2 inhibited cyclin D1 expression and G1 progression when added 12 hours after EGF stimulation, whereas depletion of EGF from the medium at this time point did not. ELISA demonstrated that EGF induced TGFα release to the medium. Cyclin D1 induction and cellular proliferation were efficiently inhibited when a neutralizing antibody to TGFα was added to the medium. This also occurred when the antibody was added 12 hours after EGF stimulation. CONCLUSION: Sustained EGF receptor activity and signaling through both Erk1/2 and PI3K pathways were necessary for proliferation. This was achieved by EGF activation of autocrine TGFα.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Hepatocytes/drug effects , Signal Transduction/drug effects , Transforming Growth Factor alpha/metabolism , Animals , Antibodies, Neutralizing/immunology , Autocrine Communication/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chromones/pharmacology , Cyclin D1/metabolism , G1 Phase , Hepatocytes/metabolism , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Transforming Growth Factor alpha/immunology
8.
J Surg Oncol ; 107(4): 393-401, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22927239

ABSTRACT

BACKGROUND: Post-operative liver regeneration may contribute to tumor recurrence. There is a theoretical need for an adjuvant therapy that can suppress tumor growth without adversely affecting post-operative liver regeneration. OBJECTIVE: To evaluate the effect of RAF inhibitor Sorafenib on cell viability and proliferation of hepatoma cells and hepatocytes in vitro and in an in vivo rat model. METHODS: Cell viability, DNA synthesis, and RAF/MAPK kinase activity in the primary hepatocyte and hepatoma cell lines were investigated after Sorafenib exposure. Sequence analysis of the B-RAF gene in hepatic cells was determined. Tumor markers were compared within the rats after 70% hepatectomy with or without daily oral gavages of Sorafenib. Liver regeneration was assessed by liver function tests and proliferation markers. RESULTS: Primary hepatocytes showed higher cell viability, proliferation rate, and stronger RAF/MAPK kinase activity compared with hepatoma cell lines. The in vivo tumor volumes, size, and metastases were significantly decreased (P < 0.05) whereas no significant change in liver regeneration related to Sorafenib exposure was found (P > 0.05). B-RAF V600E mutation was not detected neither in the hepatic cells nor untransformed hepatocytes. CONCLUSIONS: The RAF targeted inhibitor can reduce tumor growth without retarding liver regeneration in this experiment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Hepatectomy , Hepatocytes/drug effects , Liver Neoplasms/drug therapy , Liver Regeneration/drug effects , Molecular Targeted Therapy/methods , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , raf Kinases/antagonists & inhibitors , Animals , Blotting, Western , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Fluorescent Antibody Technique , Hepatectomy/adverse effects , Hepatectomy/methods , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Liver Neoplasms, Experimental/drug therapy , Neoplasm Micrometastasis , Niacinamide/pharmacology , Rats , Sorafenib
9.
J Cell Physiol ; 228(6): 1304-13, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23168795

ABSTRACT

In this study, we report a novel role of FAK as a regulator of Cdk2 in anchorage-dependent primary cultured hepatocytes. In response to EGF, we found that S-phase entry was reduced upon FAK inhibition. This correlated with decreased protein expression and nuclear accumulation of the G1/S-phase regulator Cdk2. Further, nuclear accumulation of the Cdk2 partner cyclinE, was reduced, but not its protein level. Also, protein levels of Cdk2 were inversely linked with increased expression of the Cdk2 inhibitor p27, known to be degraded in a Cdk2-dependent manner. Also, cyclinD1 was regulated by FAK, but to a lesser extent than Cdk2. To assess the mechanism in which FAK mediates Cdk2-regulation, FAK mutants were used: FAKY397F, mutated at its integrin-regulated site, and two others mutated at docking sites for Grb2-ERK-activation (FAKY925F) and for p130Cas-Rac1-activation (FAKY861F). All three sites were central for EGF-induced ERK-activity and Cdk2 expression. In addition, FAK was important for HGF-mediated proliferation, suggesting a general mechanism for anchorage-dependent growth. Moreover, growth factor-induced cell spreading, but not survival, required FAK. Hence, integrins and growth factors cooperate in anchorage-dependent signaling events leading to proliferation and motility. In conclusion, our data suggest that FAK acts as a central coordinator of integrin and growth factor-mediated S-phase entry by its ability to regulate Cdk2.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Epidermal Growth Factor/metabolism , Focal Adhesion Kinase 1/metabolism , Hepatocytes/enzymology , Active Transport, Cell Nucleus , Animals , Apoptosis , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Shape , Cells, Cultured , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Hepatocyte Growth Factor/metabolism , Hepatocytes/drug effects , Male , Mutagenesis, Site-Directed , Mutation , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , RNA Interference , Rats , Rats, Wistar , S Phase Cell Cycle Checkpoints , Signal Transduction , Transfection , Transforming Growth Factor beta1/metabolism
10.
J Biol Chem ; 287(42): 35733-35746, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22896705

ABSTRACT

The GABA transporters (GAT1, GAT2, GAT3, and BGT1) have mostly been discussed in relation to their potential roles in controlling the action of transmitter GABA in the nervous system. We have generated the first mice lacking the GAT2 (slc6a13) gene. Deletion of GAT2 (both mRNA and protein) neither affected growth, fertility, nor life span under nonchallenging rearing conditions. Immunocytochemistry showed that the GAT2 protein was predominantly expressed in the plasma membranes of periportal hepatocytes and in the basolateral membranes of proximal tubules in the renal cortex. This was validated by processing tissue from wild-type and knockout mice in parallel. Deletion of GAT2 reduced liver taurine levels by 50%, without affecting the expression of the taurine transporter TAUT. These results suggest an important role for GAT2 in taurine uptake from portal blood into liver. In support of this notion, GAT2-transfected HEK293 cells transported [(3)H]taurine. Furthermore, most of the uptake of [(3)H]GABA by cultured rat hepatocytes was due to GAT2, and this uptake was inhibited by taurine. GAT2 was not detected in brain parenchyma proper, excluding a role in GABA inactivation. It was, however, expressed in the leptomeninges and in a subpopulation of brain blood vessels. Deletion of GAT2 increased brain taurine levels by 20%, suggesting a taurine-exporting role for GAT2 in the brain.


Subject(s)
Brain/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Liver/metabolism , Taurine/metabolism , Animals , Brain/cytology , Brain Chemistry , GABA Plasma Membrane Transport Proteins/genetics , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Kidney Cortex/cytology , Kidney Cortex/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Liver/cytology , Male , Mice , Mice, Knockout , Rabbits , Rats , Rats, Wistar , Taurine/genetics
11.
J Cell Physiol ; 226(9): 2267-78, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21660950

ABSTRACT

Reactive oxygen species (ROS) function as signaling molecules mainly by reversible oxidation of redox-sensitive target proteins. ROS can be produced in response to integrin ligation and growth factor stimulation through Rac1 and its effector protein NADPH oxidase. One of the central roles of Rac1-NADPH oxidase is actin cytoskeletal rearrangement, which is essential for cell spreading and migration. Another important regulator of cell spread is focal adhesion kinase (FAK), a coordinator of integrin and growth factor signaling. Here, we propose a novel role for NADPH oxidase as a modulator of the FAK autophosphorylation site. We found that Rac1-NADPH oxidase enhanced the phosphorylation of FAK at Y397. This site regulates FAK's ability to act as a scaffold for EGF-mediated signaling, including activation of ERK. Accordingly, we found that EGF-induced activation of FAK at Y925, the following activation of ERK, and phosphorylation of FAK at the ERK-regulated S910-site depended upon NADPH oxidase. Furthermore, the inhibition of NADPH oxidase caused excessive focal adhesions, which is in accordance with ERK and FAK being modulators of focal adhesion dissociation. Our data suggest that Rac1 through NADPH oxidase is part of the signaling pathway constituted by FAK, Rac1, and ERK that regulates focal adhesion disassembly during cell spreading.


Subject(s)
Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , NADPH Oxidases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Focal Adhesions/drug effects , Focal Adhesions/enzymology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/enzymology , Male , Mice , Models, Biological , Phosphorylation/drug effects , Phosphoserine/metabolism , Rats , Rats, Wistar
12.
Liver Transpl ; 17(7): 866-74, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21542129

ABSTRACT

Liver resection and liver transplantation are the treatment modalities with the greatest potential for curing hepatocellular carcinoma (HCC). Tumor recurrence after resection for HCC is, however, a major problem, and an increased rate of recurrence after living donor transplantation versus cadaveric whole liver transplantation has been suggested. Factors involved in liver regeneration may stimulate the growth of occult tumors. The aim of this project was to test the hypothesis that a microscopic HCC tumor in the setting of partial hepatectomy would show enhanced growth and signs of increased invasiveness corresponding to the size of the liver resection. Hepatectomy was performed to various degrees in groups of Buffalo rats with the concomitant implantation of a fixed number of hepatoma cells in the remnant liver; a control group underwent only resection. After 21 days, the sizes and numbers of the tumors and the expression of alpha-fetoprotein (AFP), cyclin D1, calpain small subunit 1 (CAPNS1), CD34 (a microvessel density marker), vascular endothelial growth factor (VEGF), and vascular endothelial growth factor receptor 2 (VEGFR2) were evaluated and compared between the groups. The tumor volume and number increased significantly with the size of the partial hepatectomy (P < 0.05). The largest resections were also associated with increased hepatoma cell infiltration in the lungs and significant up-regulation of cyclin D1, AFP, CAPNS1, CD34, VEGF, and VEGFR2. The results suggest that liver regeneration after partial hepatectomy facilitates the growth and malignant transformation of microscopic HCC, and this could be significant for liver resection and partial liver transplantation strategies for HCC.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Liver Regeneration , Animals , Body Weight , Cell Line, Tumor , Disease Models, Animal , Humans , Liver Transplantation/methods , Male , Microscopy, Fluorescence , Neoplasm Invasiveness , Neoplasm Metastasis , Rats , Recurrence
13.
BMC Cell Biol ; 9: 16, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18380891

ABSTRACT

BACKGROUND: Epidermal Growth Factor Receptor (EGFR) is a key target molecule in current treatment of several neoplastic diseases. Hence, in order to develop and improve current drugs targeting EGFR signalling, an accurate understanding of how this signalling pathway is regulated is required. It has recently been demonstrated that inhibition of cAMP-dependent protein kinase (PKA) induces a ligand-independent internalization of EGFR. Cyclic-AMP-dependent protein kinase consists of a regulatory dimer bound to two catalytic subunits. RESULTS: We have investigated the effect on EGFR levels after ablating the two catalytic subunits, Calpha and Cbeta in two different models. The first model used targeted disruption of either Calpha or Cbeta in mice whereas the second model used Calpha and Cbeta RNA interference in HeLa cells. In both models we observed a significant reduction of EGFR expression at the protein but not mRNA level. CONCLUSION: Our results suggest that PKA may represent a target that when manipulated can maintain EGFR protein levels at the single cell level as well as in intact animals.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Down-Regulation/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Signal Transduction/genetics , Animals , ErbB Receptors/genetics , Gene Expression Regulation, Enzymologic/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , RNA Interference , RNA, Messenger/metabolism
14.
J Cell Physiol ; 215(3): 818-26, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18163378

ABSTRACT

Ras proteins mediate signals both via extracellular signal-regulated kinase 1 and 2 (ERK), and phosphoinositide 3-kinase (PI3K). These signals are key events in cell protection and compensatory cell growth after exposure to cell damaging and pro-apoptotic stimuli, thus maintaining homeostasis. By transfection techniques, we found that both H-Ras and K-Ras were expressed and appeared functionally active in primary hepatocytes. We compared the ability of H-Ras and K-Ras homologues to preferentially activate one of the two pathways, thereby differentially controlling cell survival and growth. We found that ectopic expression of dominant negative (DN) H-RasN17, but not DN K-RasN17, efficiently inhibited both phosphorylation and translocation of ERK to the nuclear compartment, which are prerequisites for cell cycle progression. Furthermore, ectopic expression of constitutive active (CA) H-RasV12, but not CA K-RasV12, potentiated EGF-induced proliferation. We also found that expression of CA mutants of either H-Ras or K-Ras protected hepatocytes from transforming growth factor-beta1 (TGF-beta1)-induced apoptosis. However, H-Ras-induced survival was mediated by ERK/RSK as well as by PI3K, whereas K-Ras-induced survival was mediated by PI3K only. In conclusion, H-Ras and K-Ras had differential functions in proliferation and survival of primary hepatocytes. H-Ras was the major mediator of ERK-induced proliferation and survival, whereas H-Ras and K-Ras both mediated PI3K-induced survival.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/cytology , Hepatocytes/enzymology , Oncogene Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , ras Proteins/metabolism , Animals , Apoptosis/drug effects , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Genes, Dominant , Hepatocytes/drug effects , Humans , Male , Mice , Phosphorylation/drug effects , Rats , Rats, Wistar , Transforming Growth Factor beta1/pharmacology
15.
Biochim Biophys Acta ; 1773(9): 1398-406, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17604136

ABSTRACT

Immunofluorescence analyses show that the vertebrate specific and DNA-binding protein NUCKS is distributed throughout the cytoplasm in mitotic cells and targeted to the reforming nuclei in late telophase of the cell cycle. Computer analysis of the primary structure of NUCKS revealed the presence of two regions of highly charged, basic residues, which were identified as potential nuclear localization signals (NLSs). One of these signals (NLS1) is highly conserved between the species investigated, and fits to the description of being a classical bipartite NLS. The other amino acid motif (NLS2) is less conserved and does not constitute a classical bipartite NLS consensus sequence. We have shown that each of the two putative NLSs is capable of translocating green fluorescent protein (GFP) into the nucleus. The highly conserved NLS1 is monopartite, resembling the signals of c-Myc and RanBP3. Surprisingly, a natural occurring splice variant of NUCKS lacking 40 amino acids including NLS1, is not capable of translocating a corresponding NUCKS-GFP fusion protein into the nucleus, indicating that NLS1 is the main nuclear localization signal in NUCKS. This is also confirmed by site-directed mutagenesis of the full-length protein. By GFP-immunoprecipitation and GST-pull down experiments, we show that NUCKS binds to importin alpha3 and importin alpha5 in vitro, suggesting that the nuclear targeting of NUCKS follows a receptor-mediated and energy-dependent import mechanism.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/chemistry , Nuclear Localization Signals/chemistry , Nuclear Proteins/chemistry , Phosphoproteins/chemistry , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique, Indirect , Genes, Reporter , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Luciferases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Telophase , alpha Karyopherins/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 292(1): L232-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16980378

ABSTRACT

To elucidate the role of cAMP and different cAMP-dependent protein kinases (PKA; A-kinase) in lung cell proliferation, we investigated rat alveolar type 2 cell proliferation in relation to activation or inhibition of PKA and PKA regulatory subunits (RIIalpha and RIalpha). Both the number of proliferating type 2 cells and the level of different regulatory subunits varied during 7 days of culture. The cells exhibited a distinct peak of proliferation after 5 days of culture. This proliferation peak was preceded by a rise in RIIalpha protein level. In contrast, an inverse relationship between RIalpha and type 2 cell proliferation was noted. Activation of PKA increased type 2 cell proliferation if given at peak RIIalpha expression. Furthermore, PKA inhibitors lowered the rate of proliferation only when a high RII level was observed. An antibody against the anchoring region of RIIalpha showed cell cycle-dependent binding in contrast to antibodies against other regions, possibly related to altered binding to A-kinase anchoring protein. Following activation of PKA, relocalization of RIIalpha was confirmed by immunocytochemistry. In conclusion, it appears that activation of PKA II is important in regulation of alveolar type 2 cell proliferation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/enzymology , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Cyclic AMP-Dependent Protein Kinase Type II , Enzyme Activation , Immunohistochemistry , In Vitro Techniques , Male , Rats , Rats, Inbred WKY
17.
BMC Immunol ; 7: 15, 2006 Jul 13.
Article in English | MEDLINE | ID: mdl-16839418

ABSTRACT

BACKGROUND: The activation induced T cell specific adapter protein (TSAd), encoded by SH2D2A, interacts with and modulates Lck activity. Several transcript variants of TSAd mRNA exist, but their biological significance remains unknown. Here we examined expression of SH2D2A transcripts in activated CD4+ T cells and used the SH2D2A variants as tools to identify functionally important regions of TSAd. RESULTS: TSAd was found to interact with Lck in human CD4+ T cells ex vivo. Three interaction modes of TSAd with Lck were identified. TSAd aa239-256 conferred binding to the Lck-SH3 domain, whereas one or more of the four tyrosines within aa239-334 encoded by SH2D2A exon 7 was found to confer interaction with the Lck-SH2-domain. Finally the TSAd-SH2 domain was found to interact with Lck. The SH2D2A exon 7 encoding TSAd aa 239-334 was found to harbour information essential not only for TSAd interaction with Lck, but also for TSAd modulation of Lck activity and translocation of TSAd to the nucleus. All five SH2D2A transcripts were found to be expressed in CD3 stimulated CD4+ T cells. CONCLUSION: These data show that TSAd and Lck may interact through several different domains and that Lck TSAd interaction occurs in CD4+ T cells ex vivo. Alternative splicing of exon 7 encoding aa239-334 results in loss of the majority of protein interaction motives of TSAd and yields truncated TSAd molecules with altered ability to modulate Lck activity. Whether TSAd is regulated through differential alternative splicing of the SH2D2A transcript remains to be determined.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , CD4-Positive T-Lymphocytes/immunology , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , CD4-Positive T-Lymphocytes/enzymology , Cell Nucleus/metabolism , Exons , Gene Expression , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Molecular Sequence Data , Phosphorylation , Proline/analysis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Structure-Activity Relationship , Tyrosine/metabolism , src Homology Domains
18.
Hepatology ; 42(1): 200-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15962331

ABSTRACT

Reactive oxygen species (ROS) are implicated in tissue damage causing primary hepatic dysfunction following ischemia/reperfusion injury and during inflammatory liver diseases. A potential role of extracellular signal-regulated kinase (ERK) as a mediator of survival signals during oxidative stress was investigated in primary cultures of hepatocytes exposed to ROS. Hydrogen peroxide (H(2)O(2)) induced a dose-dependent activation of ERK, which was dependent on MEK activation. The ERK activation pattern was transient compared with the ERK activation seen after stimulation with epidermal growth factor (EGF). Nuclear accumulation of ERK was found after EGF stimulation, but not after H(2)O(2) exposure. A slow import/rapid export mechanism was excluded through the use of leptomycin B, an inhibitor of nuclear export sequence-dependent nuclear export. Reduced survival of hepatocytes during ROS exposure was observed when ERK activation was inhibited. Ribosomal S6 kinase (RSK), a cytoplasmic ERK substrate involved in cell survival, was activated and located in the nucleus of H(2)O(2)-exposed hepatocytes. The activation was abolished when ERK was inhibited with U0126. In conclusion, our results indicate that activity of ERK in the cytoplasm is important for survival during oxidative stress in hepatocytes and that RSK is activated downstream of ERK. Supplementary material for this article can be found on the HEPATOLOGY website (http://www.interscience.wiley.com/jpages/0270-9139/suppmat/index.html).


Subject(s)
Extracellular Signal-Regulated MAP Kinases/drug effects , Hepatocytes/drug effects , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/drug effects , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytoplasm , Hepatocytes/metabolism , Male , Models, Animal , Oxidative Stress , Rats , Rats, Wistar , Reactive Oxygen Species , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
19.
Mol Pharmacol ; 67(5): 1581-90, 2005 May.
Article in English | MEDLINE | ID: mdl-15713850

ABSTRACT

We have previously reported that endocytic sorting of ET(A) endothelin receptors to the recycling pathway is dependent on a signal residing in the cytoplasmic carboxyl-terminal region. The aim of the present work was to characterize the carboxyl-terminal recycling motif of the ET(A) receptor. Assay of truncation mutants of the ET(A) receptor with increasing deletions of the carboxyl-terminal tail revealed that amino acids 390 to 406 contained information critical for the ability of the receptor to recycle. This peptide sequence displayed significant sequence similarity to several protein segments confirmed by X-ray crystallography to adopt antiparallel beta-strand structures (beta-finger). One of these segments was the beta-finger motif of neuronal nitric-oxide synthase reported to function as an internal PDZ (postsynaptic density-95/disc-large/zona occludens) domain-binding ligand. Based on these findings, the three-dimensional structure of the recycling motif of ET(A) receptor was predicted to attain a beta-finger conformation acting as an internal PDZ ligand. Site-directed mutagenesis at residues that would be crucial to the structural integrity of the putative beta-finger conformation or PDZ ligand function prevented recycling of the ET(A) receptor. Analysis of more than 300 G protein-coupled receptors (GPCRs) identified 35 different human GPCRs with carboxylterminal sequence patterns that fulfilled the structural criteria of an internal PDZ ligand. Among these are several receptors reported to follow a recycling pathway. In conclusion, recycling of ET(A) receptor is mediated by a motif with the structural characteristics of an internal PDZ ligand. This structural motif may represent a more general principle of endocytic sorting of GPCRs.


Subject(s)
Endocytosis/physiology , Receptor, Endothelin A/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Humans , Molecular Sequence Data , Protein Transport/physiology , Receptor, Endothelin A/genetics , Receptors, G-Protein-Coupled/genetics
20.
FEBS Lett ; 569(1-3): 207-10, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15225635

ABSTRACT

The 14-3-3 proteins are known to interact with a number of proteins involved in the regulation of cell signaling. Here, we describe an association of 14-3-3zeta with the epidermal growth factor receptor (EGFR) that is rapidly induced by EGF. The 1028-EGFR truncated mutant which lacks the cytoplasmic tail from amino acids 1029-1186 identified the binding site for 14-3-3 to be between amino acid 1028 and the receptor carboxyl terminus. Mutational deletion of serine residues 1046, 1047, 1057 and 1142 did not inhibit EGF-induced 14-3-3 association with the receptor. Immunofluorescence microscopy indicated an EGF-induced co-localization of EGFR and HA-14-3-3zeta along the plasma membrane. Our finding adds to the growing complexity of EGF receptor signaling and indicates a role for 14-3-3 proteins in EGF receptor signaling or regulation.


Subject(s)
ErbB Receptors/metabolism , Tyrosine 3-Monooxygenase/metabolism , 14-3-3 Proteins , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , HeLa Cells , Humans , Kinetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phospholipases A/metabolism , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...