Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 59(10): 6018-6032, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35835895

ABSTRACT

Lafora disease (LD) is a fatal rare neurodegenerative disorder that affects young adolescents and has no treatment yet. The hallmark of LD is the presence of polyglucosan inclusions (PGs), called Lafora bodies (LBs), in the brain and peripheral tissues. LD is caused by mutations in either EPM2A or EPM2B genes, which, respectively, encode laforin, a glucan phosphatase, and malin, an E3-ubiquitin ligase, with identical clinical features. LD knockout mouse models (Epm2a - / - and Epm2b - / -) recapitulate PG body accumulation, as in the human pathology, and display alterations in glutamatergic transmission and neuroinflammatory pathways in the brain. In this work, we show the results of four pre-clinical trials based on the modulation of glutamatergic transmission (riluzole and memantine) and anti-neuroinflammatory interventions (resveratrol and minocycline) as therapeutical strategies in an Epm2b - / - mouse model. Drugs were administered in mice from 3 to 5 months of age, corresponding to early stage of the disease, and we evaluated the beneficial effect of the drugs by in vivo behavioral phenotyping and ex vivo histopathological brain analyses. The behavioral assessment was based on a battery of anxiety, cognitive, and neurodegenerative tests and the histopathological analyses included a panel of markers regarding PG accumulation, astrogliosis, and microgliosis. Overall, the outcome of ameliorating the excessive glutamatergic neurotransmission present in Epm2b - / - mice by memantine displayed therapeutic effectiveness at the behavioral levels. Modulation of neuroinflammation by resveratrol and minocycline also showed beneficial effects at the behavioral level. Therefore, our study suggests that both therapeutical strategies could be beneficial for the treatment of LD patients. A mouse model of Lafora disease (Epm2b-/-) was used to check the putative beneficial effect of different drugs aimed to ameliorate the alterations in glutamatergic transmission and/or neuroinflammation present in the model. Drugs in blue gave a more positive outcome than the rest.


Subject(s)
Lafora Disease , Adolescent , Animals , Disease Models, Animal , Dual-Specificity Phosphatases/metabolism , Humans , Lafora Disease/genetics , Memantine , Mice , Mice, Knockout , Minocycline/pharmacology , Minocycline/therapeutic use , Myoclonic Epilepsies, Progressive , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Resveratrol , Ubiquitin-Protein Ligases/metabolism
2.
Front Mol Neurosci ; 15: 912780, 2022.
Article in English | MEDLINE | ID: mdl-35769335

ABSTRACT

Computational techniques for analyzing biological images offer a great potential to enhance our knowledge of the biological processes underlying disorders of the nervous system. Friedreich's Ataxia (FRDA) is a rare progressive neurodegenerative inherited disorder caused by the low expression of frataxin, which is a small mitochondrial protein. In FRDA cells, the lack of frataxin promotes primarily mitochondrial dysfunction, an alteration of calcium (Ca2+) homeostasis and the destabilization of the actin cytoskeleton in the neurites and growth cones of sensory neurons. In this paper, a computational multilinear algebra approach was used to analyze the dynamics of the growth cone and its function in control and FRDA neurons. Computational approach, which includes principal component analysis and a multilinear algebra method, is used to quantify the dynamics of the growth cone (GC) morphology of sensory neurons from the dorsal root ganglia (DRG) of the YG8sR humanized murine model for FRDA. It was confirmed that the dynamics and patterns of turning were aberrant in the FRDA growth cones. In addition, our data suggest that other cellular processes dependent on functional GCs such as axonal regeneration might also be affected. Semiautomated computational approaches are presented to quantify differences in GC behaviors in neurodegenerative disease. In summary, the deficiency of frataxin has an adverse effect on the formation and, most importantly, the growth cones' function in adult DRG neurons. As a result, frataxin deficient DRG neurons might lose the intrinsic capability to grow and regenerate axons properly due to the dysfunctional GCs they build.

3.
Mol Neurobiol ; 58(6): 2508-2522, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33447969

ABSTRACT

Lafora disease (LD; OMIM#274780) is a fatal rare neurodegenerative disorder characterized by generalized epileptic seizures and the presence of polyglucosan inclusions (PGs), called Lafora bodies (LBs), typically in the brain. LD is caused by mutations in two genes EPM2A or EPM2B, which encode respectively laforin, a glucan phosphatase, and malin, an E3-ubiquitin ligase. Much remains unknown about the molecular bases of LD and, unfortunately, appropriate treatment is still missing; therefore patients die within 10 years from the onset of the disease. Recently, we have identified neuroinflammation as one of the initial determinants in LD. In this work, we have investigated anti-inflammatory treatments as potential therapies in LD. With this aim, we have performed a preclinical study in an Epm2b-/- mouse model with propranolol, a ß-adrenergic antagonist, and epigallocatechin gallate (EGCG), an antioxidant from green tea extract, both of which displaying additional anti-inflammatory properties. In vivo motor and cognitive behavioral tests and ex vivo histopathological brain analyses were used as parameters to assess the therapeutic potential of propranolol and EGCG. After 2 months of treatment, we observed an improvement not only in attention defects but also in neuronal disorganization, astrogliosis, and microgliosis present in the hippocampus of Epm2b-/- mice. In general, propranolol intervention was more effective than EGCG in preventing the appearance of astrocyte and microglia reactivity. In summary, our results confirm the potential therapeutic effectiveness of the modulators of inflammation as novel treatments in Lafora disease.


Subject(s)
Brain/pathology , Inflammation/pathology , Lafora Disease/pathology , Animals , Biomarkers/metabolism , Brain/drug effects , Brain/physiopathology , Catechin/analogs & derivatives , Catechin/pharmacology , Disease Models, Animal , Gliosis/complications , Gliosis/pathology , Gliosis/physiopathology , Glucans/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inflammation/complications , Inflammation/physiopathology , Lafora Disease/complications , Lafora Disease/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity , Nerve Degeneration/complications , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/pathology , Phenotype , Propranolol/pharmacology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
4.
Sci Rep ; 10(1): 5207, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32251310

ABSTRACT

Abnormalities in actin cytoskeleton have been linked to Friedreich's ataxia (FRDA), an inherited peripheral neuropathy characterised by an early loss of neurons in dorsal root ganglia (DRG) among other clinical symptoms. Despite all efforts to date, we still do not fully understand the molecular events that contribute to the lack of sensory neurons in FRDA. We studied the adult neuronal growth cone (GC) at the cellular and molecular level to decipher the connection between frataxin and actin cytoskeleton in DRG neurons of the well-characterised YG8R Friedreich's ataxia mouse model. Immunofluorescence studies in primary cultures of DRG from YG8R mice showed neurons with fewer and smaller GCs than controls, associated with an inhibition of neurite growth. In frataxin-deficient neurons, we also observed an increase in the filamentous (F)-actin/monomeric (G)-actin ratio (F/G-actin ratio) in axons and GCs linked to dysregulation of two crucial modulators of filamentous actin turnover, cofilin-1 and the actin-related protein (ARP) 2/3 complex. We show how the activation of cofilin is due to the increase in chronophin (CIN), a cofilin-activating phosphatase. Thus cofilin emerges, for the first time, as a link between frataxin deficiency and actin cytoskeleton alterations.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cofilin 1/physiology , Friedreich Ataxia/metabolism , Growth Cones/ultrastructure , Iron-Binding Proteins/genetics , Actin Cytoskeleton/pathology , Actin-Related Protein 2-3 Complex/metabolism , Animals , Axons/chemistry , Cells, Cultured , Disease Models, Animal , Friedreich Ataxia/genetics , Ganglia, Spinal/pathology , Mice , Mice, Neurologic Mutants , Microfilament Proteins/metabolism , Mutation, Missense , Neurites/ultrastructure , Neurons/ultrastructure , Phosphoprotein Phosphatases/physiology , Phosphorylation , Phosphoserine/metabolism , Protein Processing, Post-Translational , Frataxin
5.
Neurotherapeutics ; 16(2): 432-449, 2019 04.
Article in English | MEDLINE | ID: mdl-30761510

ABSTRACT

Friedreich's ataxia (FRDA) is a neurodegenerative disorder caused by an unstable GAA repeat expansion within intron 1 of the FXN gene and characterized by peripheral neuropathy. A major feature of FRDA is frataxin deficiency with the loss of large sensory neurons of the dorsal root ganglia (DRG), namely proprioceptive neurons, undergoing dying-back neurodegeneration with progression to posterior columns of the spinal cord and cerebellar ataxia. We used isolated DRGs from a YG8R FRDA mouse model and C57BL/6J control mice for a proteomic study and a primary culture of sensory neurons from DRG to test novel pharmacological strategies. We found a decreased expression of electron transport chain (ETC) proteins, the oxidative phosphorylation (OXPHOS) system and antioxidant enzymes, confirming a clear impairment in mitochondrial function and an oxidative stress-prone phenotype. The proteomic profile also showed a decreased expression in Ca2+ signaling related proteins and G protein-coupled receptors (GPCRs). These receptors modulate intracellular cAMP/cGMP and Ca2+ levels. Treatment of frataxin-deficient sensory neurons with phosphodiesterase (PDE) inhibitors was able to restore improper cytosolic Ca2+ levels and revert the axonal dystrophy found in DRG neurons of YG8R mice. In conclusion, the present study shows the effectiveness of PDE inhibitors against axonal degeneration of sensory neurons in YG8R mice. Our findings indicate that PDE inhibitors may become a future FRDA pharmacological treatment.


Subject(s)
Axons/drug effects , Friedreich Ataxia/pathology , Ganglia, Spinal/drug effects , Phosphodiesterase Inhibitors/pharmacology , Sensory Receptor Cells/drug effects , Animals , Axons/metabolism , Axons/pathology , Calcium Signaling/drug effects , Cell Line , Disease Models, Animal , Friedreich Ataxia/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Proteomics , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology
6.
Front Mol Neurosci ; 10: 264, 2017.
Article in English | MEDLINE | ID: mdl-28912677

ABSTRACT

Friedreich's ataxia (FRDA) is a peripheral neuropathy involving a loss of proprioceptive sensory neurons. Studies of biopsies from patients suggest that axonal dysfunction precedes the death of proprioceptive neurons in a dying-back process. We observed that the deficiency of frataxin in sensory neurons of dorsal root ganglia (DRG) of the YG8R mouse model causes the formation of axonal spheroids which retain dysfunctional mitochondria, shows alterations in the cytoskeleton and it produces impairment of axonal transport and autophagic flux. The homogenous distribution of axonal spheroids along the neurites supports the existence of continues focal damages. This lead us to propose for FRDA a model of distal axonopathy based on axonal focal damages. In addition, we observed the involvement of oxidative stress and dyshomeostasis of calcium in axonal spheroid formation generating axonal injury as a primary cause of pathophysiology. Axonal spheroids may be a consequence of calcium imbalance, thus we propose the quenching or removal extracellular Ca2+ to prevent spheroids formation. In our neuronal model, treatments with BAPTA and o-phenanthroline reverted the axonal dystrophy and the mitochondrial dysmorphic parameters. These results support the hypothesis that axonal pathology is reversible in FRDA by pharmacological manipulation of intracellular Ca2+ with Ca2+ chelators or metalloprotease inhibitors, preventing Ca2+-mediated axonal injury. Thus, the modulation of Ca2+ levels may be a relevant therapeutic target to develop early axonal protection and prevent dying-back neurodegeneration.

7.
Dis Model Mech ; 9(6): 647-57, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27079523

ABSTRACT

Frataxin (FXN) deficiency causes Friedreich's ataxia (FRDA), a multisystem disorder with neurological and non-neurological symptoms. FRDA pathophysiology combines developmental and degenerative processes of dorsal root ganglia (DRG), sensory nerves, dorsal columns and other central nervous structures. A dying-back mechanism has been proposed to explain the peripheral neuropathy and neuropathology. In addition, affected individuals have non-neuronal symptoms such as diabetes mellitus or glucose intolerance. To go further in the understanding of the pathogenic mechanisms of neuropathy and diabetes associated with the disease, we have investigated the humanized mouse YG8R model of FRDA. By biochemical and histopathological studies, we observed abnormal changes involving muscle spindles, dorsal root axons and DRG neurons, but normal findings in the posterior columns and brain, which agree with the existence of a dying-back process similar to that described in individuals with FRDA. In YG8R mice, we observed a large number of degenerated axons surrounded by a sheath exhibiting enlarged adaxonal compartments or by a thin disrupted myelin sheath. Thus, both axonal damage and defects in Schwann cells might underlie the nerve pathology. In the pancreas, we found a high proportion of senescent islets of Langerhans in YG8R mice, which decreases the ß-cell number and islet mass to pathological levels, being unable to maintain normoglycemia. As a whole, these results confirm that the lack of FXN induces different pathogenic mechanisms in the nervous system and pancreas in the mouse model of FRDA: dying back of the sensory nerves, and pancreatic senescence.


Subject(s)
Aging/pathology , Axons/pathology , Friedreich Ataxia/pathology , Mutation/genetics , Pancreas/pathology , Animals , Cellular Senescence , Disease Models, Animal , Energy Metabolism , Friedreich Ataxia/genetics , Ganglia, Spinal/pathology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Muscles/pathology , Oxidation-Reduction , Peripheral Nervous System/pathology
8.
Front Cell Neurosci ; 8: 124, 2014.
Article in English | MEDLINE | ID: mdl-24860428

ABSTRACT

Friedreich ataxia is considered a neurodegenerative disorder involving both the peripheral and central nervous systems. Dorsal root ganglia (DRG) are the major target tissue structures. This neuropathy is caused by mutations in the FXN gene that encodes frataxin. Here, we investigated the mitochondrial and cell consequences of frataxin depletion in a cellular model based on frataxin silencing in SH-SY5Y human neuroblastoma cells, a cell line that has been used widely as in vitro models for studies on neurological diseases. We showed that the reduction of frataxin induced mitochondrial dysfunction due to a bioenergetic deficit and abnormal Ca(2+) homeostasis in the mitochondria that were associated with oxidative and endoplasmic reticulum stresses. The depletion of frataxin did not cause cell death but increased autophagy, which may have a cytoprotective effect against cellular insults such as oxidative stress. Frataxin silencing provoked slow cell growth associated with cellular senescence, as demonstrated by increased SA-ßgal activity and cell cycle arrest at the G1 phase. We postulate that cellular senescence might be related to a hypoplastic defect in the DRG during neurodevelopment, as suggested by necropsy studies.

9.
PLoS One ; 6(6): e20666, 2011.
Article in English | MEDLINE | ID: mdl-21687738

ABSTRACT

BACKGROUND: Friedreich's ataxia (FRDA) is a mitochondrial rare disease, which molecular origin is associated with defect in the expression of frataxin. The pathological consequences are degeneration of nervous system structures and cardiomyopathy with necrosis and fibrosis, among others. PRINCIPAL FINDINGS: Using FRDA fibroblasts we have characterized the oxidative stress status and mitochondrial biogenesis. We observed deficiency of MnSOD, increased ROS levels and low levels of ATP. Expression of PGC-1α and mtTFA was increased and the active form of the upstream signals p38 MAPK and AMPK in fibroblasts from two patients. Interestingly, the expression of energetic factors correlated with the natural history of disease of the patients, the age when skin biopsy was performed and the size of the GAA expanded alleles. Furthermore, idebenone inhibit mitochondriogenic responses in FRDA cells. CONCLUSIONS: The induction of mitochondrial biogenesis in FRDA may be a consequence of the mitochondrial impairment associated with disease evolution. The increase of ROS and the involvement of the oxidative phosphorylation may be an early event in the cell pathophysiology of frataxin deficiency, whereas increase of mitochondriogenic response might be a later phenomenon associated to the individual age and natural history of the disease, being more evident as the patient age increases and disease evolves. This is a possible explanation of heart disease in FRDA.


Subject(s)
Aging/genetics , Aging/metabolism , Fibroblasts/pathology , Friedreich Ataxia/pathology , Gene Expression Regulation , Heat-Shock Proteins/genetics , Mitochondria/metabolism , Transcription Factors/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Adolescent , Adult , Alleles , Antioxidants/pharmacology , Catalase/metabolism , Child , DNA-Binding Proteins/metabolism , Disease Progression , Energy Metabolism/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Gene Expression Regulation/drug effects , Glutathione Peroxidase/metabolism , Humans , Male , Middle Aged , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Transcription Factors/metabolism , Trinucleotide Repeats/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Mol Cell Cardiol ; 46(2): 160-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19084534

ABSTRACT

Different lines of evidence suggest that inhibition of COX-2 activity exacerbates reperfusion injury, but direct data showing beneficial effects of increased COX-2 activity are lacking. The aim of this study was to determine the effect of constitutive expression of COX-2 on cardiomyocyte tolerance to ischemia-reperfusion injury. We generated a transgenic mouse (B6D2-Tg (MHC-PTGS2)17Upme) that constitutively expresses functional human COX-2 in cardiomyocytes under the control of alpha-myosin heavy chain promoter. COX-2 expression was confirmed by immunoblotting and by increased levels of PGE(2) and PGI(2) in myocardium. Histological and echocardiographic analysis revealed no differences in the phenotype of transgenic mice (TgCOX-2) with respect to wild type (Wt) mice. Tolerance to ischemia-reperfusion injury was analysed in a Langendorff system. Reperfused TgCOX-2 hearts after 40 min of ischemia improved functional recovery (32.9+/-6.2% vs. 9.45+/-4.4%, P=0.004) and reduced cell death assessed by LDH release (43% of reduction, P<0.001) and triphenyltetrazolium staining (41% of reduction, P=0.002). Cardioprotection was not further increased by ischemic preconditioning. Pretreatment of mice with the COX-2 inhibitor DFU attenuated cardioprotection with a correlation between myocardial PGE(2) levels and the extent of cell death. NMR spectroscopy showed a marked reduction in arachidonic acid (AA) content in TgCOX-2 hearts. Both, DFU pretreatment and perfusion of TgCOX-2 hearts with AA increased myocardial AA to values similar to those measured in Wt hearts and reversed cardioprotection. We conclude that constitutive expression of COX-2 in cardiomyocytes confers a permanent cardioprotective state against reperfusion injury. Increased PGE(2) synthesis and reduced AA content could explain this effect.


Subject(s)
Cyclooxygenase 2/physiology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Animals , Arachidonic Acid/metabolism , Cyclooxygenase 2/genetics , Echocardiography , Female , Fluorescent Antibody Technique , Humans , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Transgenic
11.
Biochem J ; 416(3): 337-46, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18671671

ABSTRACT

The effect of COX (cyclo-oxygenase)-2-dependent PGs (prostaglandins) in acute liver injury has been investigated in transgenic mice that express human COX-2 in hepatocytes. We have used three well-established models of liver injury: in LPS (lipopolysaccharide) injury in D-GalN (D-galactosamine)-preconditioned mice; in the hepatitis induced by ConA (concanavalin A); and in the proliferation of hepatocytes in regenerating liver after PH (partial hepatectomy). The results from the present study demonstrate that PG synthesis in hepatocytes decreases the susceptibility to LPS/D-GalN or ConA-induced liver injury as deduced by significantly lower levels of the pro-inflammatory profile and plasmatic aminotransferases in transgenic mice, an effect suppressed by COX-2-selective inhibitors. These Tg (transgenic) animals express higher levels of anti-apoptotic proteins and exhibit activation of proteins implicated in cell survival, such as Akt and AMP kinase after injury. The resistance to LPS/D-GalN-induced liver apoptosis involves an impairment of procaspase 3 and 8 activation. Protection against ConA-induced injury implies a significant reduction in necrosis. Moreover, hepatocyte commitment to start replication is anticipated in Tg mice after PH, due to the expression of PCNA (proliferating cell nuclear antigen), cyclin D1 and E. These results show, in a genetic model, that tissue-specific COX-2-dependent PGs exert an efficient protection against acute liver injury by an antiapoptotic/antinecrotic effect and by accelerated early hepatocyte proliferation.


Subject(s)
Cyclooxygenase 2/metabolism , Hepatocytes/enzymology , Liver/enzymology , Liver/pathology , Transgenes , Animals , Apoptosis/physiology , Biomarkers/blood , Caspases/metabolism , Cell Line , Cell Proliferation , Concanavalin A/pharmacology , Cyclooxygenase 2/genetics , Cytokines/metabolism , Galactosamine/pharmacology , Gene Expression , Gene Expression Profiling , Hepatectomy , Hepatocytes/cytology , Humans , Lipopolysaccharides/pharmacology , Liver/cytology , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Prostaglandins/metabolism
12.
Hepatology ; 45(3): 631-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17326157

ABSTRACT

UNLABELLED: Cyclooxygenase-2 (COX-2) is upregulated in many cancers, and the prostanoids synthesized increase proliferation, improve angiogenesis, and inhibit apoptosis in several tissues. To explore the function of COX-2 in liver, transgenic (Tg) mice were generated containing a fusion gene (LIVhCOX-2) consisting of human COX-2 cDNA under the control of the human ApoE promoter. Six lines were developed; all of them expressed the LIVhCOX-2 transgene selectively in hepatocytes. The Tg mice exhibited a normal phenotype, and the increased levels of PGE2 found were due to the constitutively expressed COX-2. Histological analysis of different tissues and macroscopic examination of the liver showed no differences between wild-type (Wt) and Tg animals. However, Tg animals were resistant to Fas-mediated liver injury, as demonstrated by low levels of plasmatic aminotransferases, a lesser caspase-3 activation, and Bax levels and an increase in Bcl-2, Mcl-1, and xIAP proteins, when compared with the Wt animals. Moreover, the resistance to Fas-mediated apoptosis is suppressed in the presence of COX-2-selective inhibitors, which prevented prostaglandin accumulation in the liver of Tg mice. CONCLUSION: These results demonstrate that expression of COX-2-dependent prostaglandins exerted a protection against liver apoptosis.


Subject(s)
Apoptosis/physiology , Cyclooxygenase 2/metabolism , Hepatocytes/enzymology , Liver/pathology , fas Receptor/physiology , Alanine Transaminase/blood , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Caspases/metabolism , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic , Humans , Liver/enzymology , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , fas Receptor/immunology
13.
Biochem J ; 398(3): 371-80, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16800815

ABSTRACT

We have investigated the mechanism of COX-2 (cyclo-oxygenase 2)-dependent inhibition of apoptosis in liver, a key pathway underlying proliferative actions of COX-2 in liver cancers, cirrhosis, chronic hepatitis C infection and regeneration after partial hepatectomy. Stable expression of COX-2 in CHL (Chang liver) cells induced proliferation, with an increase in the proportion of cells in S-phase, but no other significant changes in cell-cycle distribution. This was associated with a marked inhibition of the apoptotic response to serum deprivation, an effect mimicked by treating empty-vector-transfected control cells (CHL-V cells) with prostaglandin E2 and prevented in COX-2-expressing cells (CHL-C cells) treated with selective inhibitors of COX-2. Serum-deprived CHL-V cells displayed several indicators of activation of intrinsic apoptosis: caspases 9 and 3 activated within 6 h and caspase 8 within 18 h, Bax expression was induced, cytochrome c was released to the cytosol, and PARP-1 [poly(ADP-ribose) polymerase 1] cleavage was evident in nuclei. COX-2 expression blocked these events, concomitant with reduced expression of p53 and promotion of Akt phosphorylation, the latter indicating activation of survival pathways. CHL cells were resistant to stimulation of the extrinsic pathway with anti-Fas antibody. Moreover, in vivo expression of GFP (green fluorescent protein)-labelled COX-2 in mice by hydrodynamics-based transient transfection conferred resistance to caspase 3 activation and apoptosis induced by stimulation of Fas.


Subject(s)
Apoptosis/physiology , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Enzymologic , Hepatocytes/enzymology , Biomarkers/metabolism , Cell Line , Cyclooxygenase 2/genetics , Humans , S Phase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...