Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Scand J Gastroenterol ; 58(3): 264-268, 2023 03.
Article in English | MEDLINE | ID: mdl-36063075

ABSTRACT

BACKGROUND AND AIMS: The recommended treatment duration of hepatitis C virus (HCV) genotype 1a (GT1a) infection with elbasvir/grazoprevir (EBR/GZR) in the presence of a high baseline viral load and resistance associated substitutions (RAS) is 16 weeks with ribavirin added. The objective of this study was to evaluate the real-world effectiveness of 12 weeks of EBR/GZR without ribavirin and regardless of baseline viral load and RAS testing. METHOD: This retrospective, observational cohort study was performed at five Norwegian hospitals that did not systematically utilize RAS testing. All adult patients with chronic HCV GT1a and compensated liver disease who had received 12 weeks of EBR/GZR without ribavirin and baseline RAS testing, were included. The primary endpoint was sustained virologic response at week 12 (SVR12), or if not available, at week 4 (SVR4). RESULTS: We included 433 patients and attained SVR data on 388. The mean age was 45.7 years (22-73 years). 67.2% were male. HIV co-infection was present in 3.8% (16/424) and cirrhosis in 4% (17/424). The viral load was >800 000 IU/mL in 55.0% (235/427) of patients. Overall SVR was achieved in 97.2% (377/388). SVR was achieved in 98.3% (169/172) of those with viral load ≤800 000 IU/mL and in 96.2% (202/210) of those with viral load >800 000 IU/mL. CONCLUSION: We observed high SVR rates among patients with HCV GT1a infection treated with EBR/GZR for 12 weeks without ribavirin, with no regard to baseline viral load and no RAS testing.


Subject(s)
Hepatitis C, Chronic , Hepatitis C , Adult , Humans , Male , Middle Aged , Female , Ribavirin/therapeutic use , Antiviral Agents/therapeutic use , Hepacivirus/genetics , Retrospective Studies , Drug Therapy, Combination , Hepatitis C/drug therapy , Hepatitis C/complications , Hepatitis C, Chronic/complications , Genotype
2.
Environ Int ; 74: 170-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25454234

ABSTRACT

The purposes of this review are to assess the human exposure and human and experimental evidence for adverse effects of brominated flame-retardants (BFRs) with specific focus on intake from seafood. The leakage of BFRs from consumer products leads to exposure of humans from fetal life to adulthood. Fish and fish products contain the highest levels of BFRs and dominate the dietary intake of frequent fish eaters in Europe, while meat, followed by seafood and dairy products accounted for the highest US dietary intake. House dust is also reported as an important source of exposure for children as well as adults. The levels of BFRs in the general North American populations are higher than those in Europe and Japan and the highest levels are detected in infants and toddlers. The daily intake via breast milk exceeds the RfD in 10% of US infants. BFRs including PBDEs, HBCDs and TBBP-A have induced endocrine-, reproductive- and behavior effects in laboratory animals. Furthermore, recent human epidemiological data demonstrated association between exposure to BFRs and similar adverse effects as observed in animal studies. Fish including farmed fish and crude fish oil for human consumption may contain substantial levels of BFRs and infants and toddlers consuming these products on a daily basis may exceed the tolerable daily intake suggesting that fish and fish oil alone represent a risk to human health. This intake comes in addition to exposure from other sources (breast milk, other food, house dust). Because potential harmful concentrations of BFRs and other toxicants occur in fish and fish products, research on a wider range of products is warranted, to assess health hazard related to the contamination of fish and fish products for human consumption.


Subject(s)
Environmental Exposure , Environmental Pollutants/toxicity , Flame Retardants/toxicity , Hydrocarbons, Brominated/toxicity , Adult , Animals , Child , Child, Preschool , Environmental Pollutants/analysis , Environmental Pollutants/chemistry , Environmental Pollutants/pharmacokinetics , Female , Fishes , Flame Retardants/analysis , Flame Retardants/pharmacokinetics , Humans , Hydrocarbons, Brominated/analysis , Hydrocarbons, Brominated/chemistry , Hydrocarbons, Brominated/pharmacokinetics , Infant , Male , Mice , Rats , Risk Assessment , Seafood/analysis
3.
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
4.
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
5.
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
6.
FASEB J ; 22(2): 466-76, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17928366

ABSTRACT

In this study, we provide novel insight into the mechanism of how ERK2 can be sorted to different intracellular compartments and thereby mediate different responses. MEK1-activated ERK2 accumulated in the nucleus and induced proliferation. Conversely, MEK2-activated ERK2 was retained in the cytoplasm and allowed survival. Localization was a determinant for ERK2 functions since MEK1 switched from providing proliferation to be a mediator of survival when ERK2 was routed to the cytoplasm by the attachment of a nuclear export site. MEK1-mediated ERK2 nuclear translocation and proliferation were shown to depend on phosphorylation of S298 and T292 sites in the MEK1 proline-rich domain. These sites are phosphorylated on cellular adhesion in MEK1 but not MEK2. Whereas p21-activated kinase phosphorylates S298 and thus enhances the MEK1-ERK2 association, ERK2 phosphorylates T292, leading to release of active ERK2 from MEK1. On the basis of these results, we propose that the requirement of adhesion for cells to proliferate in response to growth factors, in part, may be explained by the MEK1 S298/T292 control of ERK2 nuclear translocation. In addition, we suggest that ERK2 intracellular localization determines whether growth factors mediate proliferation or survival and that the sorting occurs in an adhesion-dependent manner.


Subject(s)
MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , DNA/biosynthesis , Gene Expression Regulation, Enzymologic , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Male , Mitogen-Activated Protein Kinase 1/genetics , Mutation/genetics , Phosphoserine/metabolism , Phosphothreonine/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta/pharmacology
7.
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
8.
Brain Res ; 984(1-2): 111-21, 2003 Sep 12.
Article in English | MEDLINE | ID: mdl-12932845

ABSTRACT

Many recent reports on internucleosomal DNA fragments have appeared, however, little is known about the mechanisms of the generation of their upstream high molecular weight (HMW) fragments. Caspases are a family of proteases with important functions in the execution of apoptotic cell death. The caspase-sensitivity of the formation of HMW fragments was therefore investigated using a specific caspase-3 inhibitor (Ac-DEVD-cmk) and a general caspase inhibitor (boc-D-fmk). Apoptosis inducing factor (AIF) can translocate to the nucleus and generate HMW fragments independently of caspase. Cultures of cerebellar granule neurons (CGNs) were therefore exposed to glutamate (100 micro M) or deprived of potassium and serum to induce apoptosis, or treated with a high concentration of calcium ionophore A23187 (1 micro M) to induce necrosis. Fragmentation of DNA into two classes of HMW fragments (>680 and 50-300 kbp) was observed after treatment with glutamate or A23187. Traces of approximately 50-kbp fragments were detectable after the K(+)/serum-deprivation. The amount of >680-kbp HMW fragments increased (i.e. their further degradation was inhibited) and cell death was reduced in the presence of Ac-DEVD-cmk or boc-D-fmk following glutamate treatment. Only boc-D-fmk treatment resulted in a similar accumulation of >680-kbp HMW fragments and reduced cell death after K(+)/serum-deprivation. No such changes were observed with caspase inhibitors after A23187 treatment. AIF redistribution was observed following glutamate treatment and K(+)/serum-deprivation. Thus, even in a simple cell culture of CGNs, HMW fragments are formed by diverse mechanisms: the degradation of DNA may be sensitive to different caspases or be caspase and AIF independent.


Subject(s)
Caspases/metabolism , Cerebellum/metabolism , DNA Fragmentation/physiology , DNA/metabolism , Neurons/metabolism , Animals , Caspase Inhibitors , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebellum/drug effects , DNA Fragmentation/drug effects , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...