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











Publication year range
1.
Sci Rep ; 9(1): 16665, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31723151

ABSTRACT

Echocardiography has become an indispensable tool for the study of heart performance, improving the monitoring of individuals with cardiac diseases. Diverse genetic factors associated with echocardiographic measures have been previously reported. The impact of several apoptotic genes in heart development identified in experimental models prompted us to assess their potential association with human cardiac function. This study aimed at investigating the possible association of variants of apoptotic genes with echocardiographic traits and to identify new genetic markers associated with cardiac function. Genome wide data from different studies were obtained from public repositories. After quality control and imputation, a meta-analysis of individual association study results was performed. Our results confirmed the role of caspases and other apoptosis related genes with cardiac phenotypes. Moreover, enrichment analysis showed an over-representation of genes, including some apoptotic regulators, associated with Alzheimer's disease. We further explored this unexpected observation which was confirmed by genetic correlation analyses. Our findings show the association of apoptotic gene variants with echocardiographic indicators of heart function and reveal a novel potential genetic link between echocardiographic measures in healthy populations and cognitive decline later on in life. These findings may have important implications for preventative strategies combating Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Genetic Markers , Genome-Wide Association Study/methods , Heart Diseases/genetics , Heart Diseases/physiopathology , Polymorphism, Single Nucleotide , Adolescent , Adult , Female , Genetic Loci , Genetic Predisposition to Disease , Humans , Male , Meta-Analysis as Topic , Phenotype , Young Adult
2.
Oncogene ; 35(39): 5179-90, 2016 09 29.
Article in English | MEDLINE | ID: mdl-26996667

ABSTRACT

Neuroblastoma (NB) is a neoplasm of the sympathetic nervous system, and is the most common solid tumor of infancy. NBs are very heterogeneous, with a clinical course ranging from spontaneous regression to resistance to all current forms of treatment. High-risk patients need intense chemotherapy, and only 30-40% will be cured. Relapsed or metastatic tumors acquire multi-drug resistance, raising the need for alternative treatments. Owing to the diverse mechanisms that are responsible of NB chemoresistance, we aimed to target epigenetic factors that control multiple pathways to bypass therapy resistance. We found that the SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4/BRG1) was consistently upregulated in advanced stages of NB, with high BRG1 levels being indicative of poor outcome. Loss-of-function experiments in vitro and in vivo showed that BRG1 is essential for the proliferation of NB cells. Furthermore, whole-genome transcriptome analysis revealed that BRG1 controls the expression of key elements of oncogenic pathways such as PI3K/AKT and BCL2, which offers a promising new combination therapy for high-risk NB.


Subject(s)
Cell Survival/genetics , DNA Helicases/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcriptome/genetics , Cell Death/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/genetics
3.
Cell Death Dis ; 6: e1639, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25675299

ABSTRACT

The brains of patients with Alzheimer's disease (AD) present elevated levels of tumor necrosis factor-α (TNFα), a cytokine that has a dual function in neuronal cells. On one hand, TNFα can activate neuronal apoptosis, and on the other hand, it can protect these cells against amyloid-ß (Aß) toxicity. Given the dual behavior of this molecule, there is some controversy regarding its contribution to the pathogenesis of AD. Here we examined the relevance of the long form of Fas apoptotic inhibitory molecule (FAIM) protein, FAIM-L, in regulating the dual function of TNFα. We detected that FAIM-L was reduced in the hippocampi of patients with AD. We also observed that the entorhinal and hippocampal cortex of a mouse model of AD (PS1(M146L)xAPP(751sl)) showed a reduction in this protein before the onset of neurodegeneration. Notably, cultured neurons treated with the cortical soluble fractions of these animals showed a decrease in endogenous FAIM-L, an effect that is mimicked by the treatment with Aß-derived diffusible ligands (ADDLs). The reduction in the expression of FAIM-L is associated with the progression of the neurodegeneration by changing the inflammatory response mediated by TNFα in neurons. In this sense, we also demonstrate that the protection afforded by TNFα against Aß toxicity ceases when endogenous FAIM-L is reduced by short hairpin RNA (shRNA) or by treatment with ADDLs. All together, these results support the notion that levels of FAIM-L contribute to determine the protective or deleterious effect of TNFα in neuronal cells.


Subject(s)
Amyloid beta-Peptides/pharmacology , Tumor Necrosis Factors/pharmacology , Animals , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , Humans , In Vitro Techniques , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , PC12 Cells , Rats
4.
Cell Death Dis ; 5: e1435, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25275596

ABSTRACT

Glioblastoma multiforme is resistant to conventional anti-tumoral treatments due to its infiltrative nature and capability of relapse; therefore, research efforts focus on characterizing gliomagenesis and identifying molecular targets useful on therapy. New therapeutic strategies are being tested in patients, such as Histone deacetylase inhibitors (HDACi) either alone or in combination with other therapies. Here two HDACi included in clinical trials have been tested, suberanilohydroxamic acid (SAHA) and valproic acid (VPA), to characterize their effects on glioma cell growth in vitro and to determine the molecular changes that promote cancer cell death. We found that both HDACi reduce glioma cell viability, proliferation and clonogenicity. They have multiple effects, such as inducing the production of reactive oxygen species (ROS) and activating the mitochondrial apoptotic pathway, nevertheless cell death is not prevented by the pan-caspase inhibitor Q-VD-OPh. Importantly, we found that HDACi alter cell cycle progression by decreasing the expression of G2 checkpoint kinases Wee1 and checkpoint kinase 1 (Chk1). In addition, HDACi reduce the expression of proteins involved in DNA repair (Rad51), mitotic spindle formation (TPX2) and chromosome segregation (Survivin) in glioma cells and in human glioblastoma multiforme primary cultures. Therefore, HDACi treatment causes glioma cell entry into mitosis before DNA damage could be repaired and to the formation of an aberrant mitotic spindle that results in glioma cell death through mitotic catastrophe-induced apoptosis.


Subject(s)
G2 Phase Cell Cycle Checkpoints/drug effects , Glioma/physiopathology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Mitosis/drug effects , Valproic Acid/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Checkpoint Kinase 1 , Glioma/drug therapy , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Vorinostat
5.
Cell Death Dis ; 5: e1401, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25188511

ABSTRACT

Neuroblastoma (NBL) is the most common solid tumor in infants and accounts for 15% of all pediatric cancer deaths. Several risk factors predict NBL outcome: age at the time of diagnosis, stage, chromosome alterations and MYCN (V-Myc Avian Myelocytomatosis Viral Oncogene Neuroblastoma-Derived Homolog) amplification, which characterizes the subset of the most aggressive NBLs with an overall survival below 30%. MYCN-amplified tumors develop exceptional chemoresistance and metastatic capacity. These properties have been linked to defects in the apoptotic machinery, either by silencing components of the extrinsic apoptotic pathway (e.g. caspase-8) or by overexpression of antiapoptotic regulators (e.g. Bcl-2, Mcl-1 or FLIP). Very little is known on the implication of death receptors and their antagonists in NBL. In this work, the expression levels of several death receptor antagonists were analyzed in multiple human NBL data sets. We report that Lifeguard (LFG/FAIM2 (Fas apoptosis inhibitory molecule 2)/NMP35) is downregulated in the most aggressive and undifferentiated tumors. Intringuingly, although LFG has been initially characterized as an antiapoptotic protein, we have found a new association with NBL differentiation. Moreover, LFG repression resulted in reduced cell adhesion, increased sphere growth and enhanced migration, thus conferring a higher metastatic capacity to NBL cells. Furthermore, LFG expression was found to be directly repressed by MYCN at the transcriptional level. Our data, which support a new functional role for a hitherto undiscovered MYCN target, provide a new link between MYCN overexpression and increased NBL metastatic properties.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Membrane Proteins/metabolism , Neuroblastoma/pathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Animals , Anti-Bacterial Agents/toxicity , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Cell Adhesion , Cell Differentiation , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Doxycycline/toxicity , Female , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Nude , N-Myc Proto-Oncogene Protein , Neoplasm Metastasis , Neoplasm Staging , Neuroblastoma/metabolism , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Death Domain/antagonists & inhibitors , Receptors, Death Domain/metabolism , Transplantation, Heterologous , Tretinoin/pharmacology , Up-Regulation/drug effects
6.
Cell Death Dis ; 4: e602, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23598414

ABSTRACT

Neurotrophins are involved in many crucial cellular functions, including neurite outgrowth, synapse formation, and plasticity. Although these events have long been known, the molecular determinants underlying neuritogenesis have not been fully characterized. Ack1 (activated Cdc42-associated tyrosine kinase) is a non-receptor tyrosine kinase that is highly expressed in the brain. Here, we demonstrate that Ack1 is a molecular constituent of neurotrophin signaling cascades in neurons and PC12 cells. We report that Ack1 interacts with Trk receptors and becomes tyrosine phosphorylated and its kinase activity is increased in response to neurotrophins. Moreover, our data indicate that Ack1 acts upstream of the Akt and MAPK pathways. We show that Ack1 overexpression induces neuritic outgrowth and promotes branching in neurotrophin-treated neuronal cells, whereas the expression of Ack1 dominant negatives or short-hairpin RNAs counteract neurotrophin-stimulated differentiation. Our results identify Ack1 as a novel regulator of neurotrophin-mediated events in primary neurons and in PC12 cells.


Subject(s)
Nerve Growth Factors/metabolism , Neurites/physiology , Protein-Tyrosine Kinases/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurogenesis/drug effects , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Rats , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Signal Transduction/drug effects
7.
Cell Death Dis ; 4: e493, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23412386

ABSTRACT

Activation of tumor necrosis factor receptor-1 can trigger survival or apoptosis pathways. In many cellular models, including the neuronal cell model PC12, it has been demonstrated that inhibition of protein synthesis is sufficient to render cells sensitive to apoptosis induced by TNFα. The survival effect is linked to the translocation of the transcription factor nuclear factor-kappa B (NF-κB) to the nucleus and activation of survival-related genes such as FLICE-like inhibitory protein long form (FLIP-L) or IAPs. Nonetheless, we previously reported an NF-κB-independent contribution of Bcl-xL to cell survival after TNFα treatment. Here, we demonstrate that NF-κB-induced increase in FLIP-L expression levels is essential for mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK) activation. We demonstrate that FLIP-L behaves as a Raf-1 activator through both protein-protein interaction and Raf-1 kinase activation, without the requirement of the classical Ras activation. Importantly, prevention of FLIP-L increase by NF-κB inhibition or knockdown of endogenous FLIP-L blocks MAPK/ERK activation after TNFα treatment. From a functional point of view, we show that inhibition of the MAPK/ERK pathway and the NF-κB pathway are equally relevant to render PC12 cells sensitive to cell death induced by TNFα. Apoptosis induced by TNFα under these conditions is dependent on jun nuclear kinase1/2 JNK1/2-dependent Bim upregulation. Therefore, we report a previously undescribed and essential role for MAPK/ERK activation by FLIP-L in the decision between cell survival and apoptosis upon TNFα stimulation.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Nucleus/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , NF-kappa B/metabolism , PC12 Cells , Protein Interaction Maps , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , ras Proteins/metabolism
8.
Cell Death Dis ; 2: e149, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21525936

ABSTRACT

Disturbance of calcium homeostasis and accumulation of misfolded proteins in the endoplasmic reticulum (ER) are considered contributory components of cell death after ischemia. However, the signal-transducing events that are activated by ER stress after cerebral ischemia are incompletely understood. In this study, we show that caspase-12 and the PERK and IRE pathways are activated following oxygen-glucose deprivation (OGD) of mixed cortical cultures or neonatal hypoxia-ischemia (HI). Activation of PERK led to a transient phosphorylation of eIF2α, an increase in ATF4 levels and the induction of gadd34 (a subunit of an eIF2α-directed phosphatase). Interestingly, the upregulation of ATF4 did not lead to an increase in the levels of CHOP. Additionally, IRE1 activation was mediated by the increase in the processed form of xbp1, which would be responsible for the observed expression of edem2 and the increased levels of the chaperones GRP78 and GRP94. We were also able to detect caspase-12 proteolysis after HI or OGD. Processing of procaspase-12 was mediated by NMDA receptor and calpain activation. Moreover, our data suggest that caspase-12 activation is independent of the unfolded protein response activated by ER stress.


Subject(s)
Caspase 12/metabolism , Cerebral Cortex/cytology , Endoplasmic Reticulum/physiology , Glucose/deficiency , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/metabolism , Animals , Animals, Newborn , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Calpain/metabolism , Cell Culture Techniques , Cell Hypoxia , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Membrane Proteins/genetics , Protein Biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction , Stress, Physiological , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , X-Box Binding Protein 1
9.
Cell Death Differ ; 16(11): 1460-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19590510

ABSTRACT

Cardiac morphologic abnormalities in mice deficient for key regulators of the caspase-dependent signaling underscored its role in heart development. However, the mechanisms regulating apoptotic gene expression in the developing heart are unknown. As polypyrimidine tract binding proteins (PTB) determine gene isoform expression during myoblast differentiation and contribute to Apaf-1 translation in cell lines, we investigated whether PTB regulate apoptotic gene expression in differentiating cardiomyocytes. Our results show that PTB are expressed in the embryonic heart and are silenced during development, coinciding with a reduction in the expression of apoptotic genes. Overexpression of PTB in postnatal cardiomyocytes, which express low levels of PTB and apoptotic genes, induced an increase in the amount of pro-apoptotic proteins without affecting abundance of their respective transcripts. Translation of the reporter gene Firefly Luciferase preceded by the 5'-untranslated region of Apaf-1 or Caspase-3 was enhanced by PTB in cardiomyocytes. PTB silencing in fibroblasts induced a decrease of apoptotic protein levels. PTB overexpression in cardiomyocytes induced caspase activity and caspase-dependent DNA fragmentation during ischemia, which is otherwise caspase-independent in differentiated cardiomyocytes. Our results show that PTB contribute to apoptotic gene expression and modulate the susceptibility to caspase activation in differentiating rat cardiomyocytes.


Subject(s)
Apoptosis , Caspase 3/metabolism , Myocytes, Cardiac/cytology , Polypyrimidine Tract-Binding Protein/metabolism , 5' Untranslated Regions , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 3/genetics , Cell Differentiation , DNA Fragmentation , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Mice , Polypyrimidine Tract-Binding Protein/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Signal Transduction
10.
Cell Death Differ ; 15(9): 1510-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18497757

ABSTRACT

We analyzed the survival responses and downstream signaling elicited by GDNF on sympathetic neurons from different Ret knockin mice. Lack of tyrosine 1062, a multidocking site in Ret, completely prevented GDNF-mediated survival. Importantly, lack of tyrosine 981, although abrogating Akt phosphorylation, had no effect on neuronal survival, indicating that the PI 3-K/Akt pathway is not necessary for survival of sympathetic neurons. In contrast, silencing of B-Raf completely prevented not only GDNF-mediated but also NGF-mediated cell survival, independently of MEK-1/2. We identified IKKs as the main effectors of the protective effects of B-Raf. First, B-Raf interacted with and activated IKKs. Second, knockdown of IKKs reversed the protection afforded by a constitutively active form of B-Raf. Third, knockdown of IKKs prevented both NGF- and GDNF-mediated survival. In conclusion, our data delineate a novel survival pathway for sympathetic neurons linking B-Raf to IKKs, independently of both PI 3-K and MEK-1/2 pathways.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , I-kappa B Kinase/physiology , Neurons/enzymology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-ret/metabolism , Animals , Cell Size , Cell Survival , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, Mutant Strains , Mutation , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/chemistry , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction , Sympathetic Nervous System/cytology , Tyrosine/genetics
11.
Mol Pharmacol ; 71(3): 736-43, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17172466

ABSTRACT

Malonate, an inhibitor of mitochondrial complex II, is a widely used toxin to study neurodegeneration in Huntington's disease and ischemic stroke. We have shown previously that malonate increased reactive oxygen species (ROS) production in human SH-SY5Y neuroblastoma cells, leading to oxidative stress, cytochrome c release, and apoptotic cell death. Expression of a green fluorescent protein-Bax fusion protein in SH-SY5Y neuroblastoma cells demonstrated a Bax redistribution from the cytosol to mitochondria after 12 to 24 h of malonate treatment that coincided with mitochondrial potential collapse and chromatin condensation. Inhibition of Bax translocation using furosemide, as well as Bax gene deletion, afforded significant protection against malonate-induced apoptosis. Further experiments revealed that malonate induced a prominent increase in the level of activated p38 mitogen-activated protein (MAP) kinase and that treatment with the p38 MAP kinase inhibitor SKF86002 potently blocked malonate-induced Bax translocation and apoptosis. Treatment with vitamin E diminished ROS production, reduced the activation status of p38 MAP kinase, inhibited Bax translocation, and protected against malonate-induced apoptosis. Our data suggest that malonate-induced ROS production and subsequent p38 MAP kinase activation mediates the activation of the pro-apoptotic Bax protein to induce mitochondrial membrane permeabilization and neuronal apoptosis.


Subject(s)
Apoptosis/drug effects , Cytochromes c/metabolism , Malonates/pharmacology , Mitochondria/drug effects , Reactive Oxygen Species , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured , Malondialdehyde/analysis , Mitochondria/metabolism , Protein Transport/drug effects , Rats
12.
Cell Death Differ ; 10(9): 977-86, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12934072

ABSTRACT

Apoptosis plays a role in cardiomyocyte death in several cardiovascular disorders. Here, we show that primary postnatal cardiomyocytes did not die upon activation of the intrinsic (cytochrome c-dependent) apoptotic pathway. Release of cytochrome c from mitochondria to the cytosol occurred, but did not activate the effector phase of apoptosis. Myocardial cells did not express apoptotic protease-activating factor-1 (Apaf-1), the allosteric activator of caspase-9 acting downstream of cytochrome c release. Forced expression of Apaf-1 restored the competence to complete the cytochrome c-induced apoptotic program and this effect was prevented by overexpression of Bcl-X(L). However, cardiomyocytes were able to enter the apoptotic program when it was initiated by activation of death receptors, as observed during serum deprivation and metabolic inhibition. Our results indicate that regulation of Apaf-1 expression may be a new regulatory mechanism developed in postmitotic cells in order to prevent irreversible commitment to die after release of cytochrome c.


Subject(s)
Apoptosis , Cytochromes c/metabolism , Myocytes, Cardiac/metabolism , Proteins/metabolism , Animals , Apoptotic Protease-Activating Factor 1 , Biological Transport/drug effects , Cells, Cultured , Culture Media, Serum-Free , DNA Fragmentation , Glucose/metabolism , Mitochondria/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Proteins/analysis , Rats , Rats, Sprague-Dawley , Staurosporine/toxicity
13.
Neuropharmacology ; 44(4): 482-92, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12646285

ABSTRACT

Opioid peptides and alkaloids exert their effects via G protein-coupled receptors (GPCRs). It has been shown that, in addition to trophic factors, some GPCRs are able to activate the phosphatidylinositol 3-kinase/Akt (PI 3-K/Akt) signal transduction pathway, thus leading to cell survival. The aim of this study was to test whether activation of mu-opioid receptors has protective effects on serum withdrawal-induced cell death and to study the possible implication of PI 3-K in this process. In SH-SY5Y neuroblastoma cells fully differentiated by exposure to retinoic acid for five days, the enkephalin derivative selective mu-agonist DAMGO (0.1-2 microM) and the alkaloid morphine (0.1-10 microM) promoted cell survival after serum deprivation (MTT and trypan blue exclusion assays), without inducing cell proliferation. These effects were fully reversed by naloxone, by the selective mu-antagonist beta-funaltrexamine (beta-FNA) and also by the specific PI 3-K inhibitor LY294002. The two agonists stimulated Akt phosphorylation and the effect was also abolished by beta-FNA and by LY294002. In mouse primary cortical neurons, DAMGO reduced the percentage of apoptosis after 6, 12, 24 and 48 h of serum withdrawal; as determined by Hoechst staining. This effect was blocked by beta-FNA, by pre-treatment with pertussis toxin and by LY294002. DAMGO also stimulated Akt phosphorylation via PI 3-K in this primary neuronal culture. Together, these results indicate that stimulation of the mu-opioid receptor promotes neuronal survival in a G(i/o)-linked, PI 3-K-dependent signaling cascade and suggest that Akt may be a key downstream kinase involved in this anti-apoptotic effect.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/cytology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Receptors, Opioid, mu/agonists , Cell Differentiation , Cell Survival/drug effects , Cells, Cultured , Culture Media, Serum-Free , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , In Vitro Techniques , Morphine/pharmacology , Neurons/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Opioid, mu/antagonists & inhibitors , Signal Transduction/drug effects , Tretinoin/pharmacology , Tumor Cells, Cultured
14.
Cell Death Differ ; 9(10): 1078-89, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12232796

ABSTRACT

Caspases are a large family of cysteine proteases that play an essential role as effectors of apoptosis in metazoans. Thirteen different caspases have been identified in vertebrates so far, and their function in apoptotic or inflammatory responses is well documented. We have taken advantage of the broadly accepted condition of amphioxus (Cephalochordata, Branchiostoma floridae) as the closest living relative to vertebrates to study the molecular evolution of caspases. Here we report for the first time the pattern of programmed cell death during development of cephalochordates. We also describe the isolation and functional characterisation of the first caspase related gene in amphioxus, which we named AmphiCASP-3/7. The amphioxus caspase is expressed throughout development, from the gastrula to larva stage. AmphiCASP-3/7 induced cell death when ectopically expressed in human HEK 293T cells, and the recombinant protein was inhibited by DEVD peptides. AmphiCASP-3/7 reflects the primitive condition of the executor vertebrates caspases -3 and -7, prior to vertebrate specific duplication. Interestingly, AmphiCASP-3/7 is functionally closer to vertebrate caspase-7, as shown by substrate specificity both in vitro and in MCF7 cells. Our phylogenetic and functional data help in drawing the evolutionary history of caspases, and illustrates an example of acquisition in vertebrates of novel functional properties after gene duplication.


Subject(s)
Apoptosis/genetics , Caspases/isolation & purification , Chordata, Nonvertebrate/enzymology , Animals , Caspase 3 , Caspase 7 , Caspases/deficiency , Caspases/genetics , Chordata, Nonvertebrate/embryology , Chordata, Nonvertebrate/growth & development , DNA, Complementary/analysis , DNA, Complementary/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/enzymology , Embryo, Nonmammalian , Evolution, Molecular , Female , Gene Expression Regulation, Enzymologic/physiology , Humans , Larva/cytology , Larva/enzymology , Larva/growth & development , Male , Molecular Sequence Data , Phenotype , Phylogeny , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
15.
Mol Cell Neurosci ; 18(6): 619-31, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11749038

ABSTRACT

To determine which intracellular pathways mediate the survival effects of ciliary neurotrophic factor and cardiotrophin-1 cytokines on motoneurons, we studied the activation of the Jak/STAT, the PI 3-kinase/Akt, and the ERK pathways. At shorter time points, cytokines induced the activation of STAT3 and ERK, but not PI 3-kinase. Jak3 inhibitor suppressed cytokine- and muscle extract-induced survival. In contrast, PD 98059, a MEK inhibitor, was not able to prevent cytokine-induced survival, demonstrating that ERK is not involved. Surprisingly, the PI 3-kinase inhibitor LY 294002 prevented the survival-promoting effects of cytokines. When assays of PI 3-kinase activity were performed at later stages following cytokine treatment a significant increase was observed compared to control cultures. This delayed increase of activity could be completely prevented by treatment with protein synthesis or Jak3 inhibitors. Collectively, these results demonstrate that cytokines induce motoneuron survival through a PI 3-kinase activation requiring de novo protein synthesis dependent on Jak pathway.


Subject(s)
Cell Survival/physiology , Cytokines/metabolism , Motor Neurons/metabolism , Nerve Growth Factors , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Spinal Cord/embryology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Cytokines/pharmacology , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Glial Cell Line-Derived Neurotrophic Factor , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Immunohistochemistry , Janus Kinase 1 , Janus Kinase 3 , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Motor Neurons/cytology , Motor Neurons/drug effects , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/growth & development , Trans-Activators/drug effects , Trans-Activators/metabolism
16.
J Cell Biol ; 154(3): 585-97, 2001 Aug 06.
Article in English | MEDLINE | ID: mdl-11489918

ABSTRACT

It has been reported that phosphoinositide 3-kinase (PI 3-kinase) and its downstream target, protein kinase B (PKB), play a central role in the signaling of cell survival triggered by neurotrophins (NTs). In this report, we have analyzed the involvement of Ca2+ and calmodulin (CaM) in the activation of the PKB induced by NTs. We have found that reduction of intracellular Ca2+ concentration or functional blockade of CaM abolished NGF-induced activation of PKB in PC12 cells. Similar results were obtained in cultures of chicken spinal cord motoneurons treated with brain-derived neurotrophic factor (BDNF). Moreover, CaM inhibition prevented the cell survival triggered by NGF or BDNF. This effect was counteracted by the transient expression of constitutive active forms of the PKB, indicating that CaM regulates NT-induced cell survival through the activation of the PKB. We have investigated the mechanisms whereby CaM regulates the activation of the PKB, and we have found that CaM was necessary for the proper generation and/or accumulation of the products of the PI 3-kinase in intact cells.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Calmodulin/metabolism , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/enzymology , Protein Serine-Threonine Kinases , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/antagonists & inhibitors , Cell Membrane/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Chelating Agents/pharmacology , Chromones/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , Morpholines/pharmacology , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/metabolism , Signal Transduction/physiology , Sulfonamides/pharmacology
17.
J Biol Chem ; 276(25): 22323-31, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11294834

ABSTRACT

Caspase-activated DNase is responsible for the oligonucleosomal DNA degradation during apoptosis. DNA degradation is thought to be important for multicellular organisms to prevent oncogenic transformation or as a mechanism of viral defense. It has been reported that certain cells, including some neuroblastoma cell lines such as IMR-5, enter apoptosis without digesting DNA in such a way. We have analyzed the causes for the absence of DNA laddering in staurosporine-treated IMR-5 cells, and we have found that most of the molecular mechanisms controlling apoptosis are well preserved in this cell line. These include degradation of substrates for caspases, blockade of cell death by antiapoptotic genes such as Bcl-2 or Bcl-X(L), or normal levels and adequate activation of caspase-3. Moreover, these cells display normal levels of caspase-activated DNase and its inhibitory protein, inhibitor of caspase-activated DNase, and their cDNA sequences are identical to those reported previously. Nevertheless, IMR-5 cells lose caspase-activated DNase during apoptosis and recover their ability to degrade DNA when human recombinant caspase-activated DNase is overexpressed. Our results lead to the conclusion that caspase-activated DNase is processed during apoptosis of IMR-5 cells, making these cells a good model to study the relevance of this endonuclease in physiological or pathological conditions.


Subject(s)
Apoptosis , Deoxyribonucleases/metabolism , Neuroblastoma/pathology , Nucleosomes/metabolism , Base Sequence , Chromatin/metabolism , DNA Primers , DNA, Neoplasm/metabolism , Humans , Hydrolysis , Tumor Cells, Cultured
18.
J Neurosci ; 21(5): 1464-72, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222636

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs), consisting of GDNF, neurturin, persephin, and artemin, signal via a multicomponent complex composed of Ret tyrosine kinase and the glycosyl-phosphatidylinositol (GPI)-anchored coreceptors GFRalpha1-alpha4. In previous work we have demonstrated that the localization of Ret to membrane microdomains known as lipid rafts is essential for GDNF-induced downstream signaling, differentiation, and neuronal survival. Moreover, we have found that Ret interacts with members of the Src family kinases (SFK) only when it is localized to these microdomains. In the present work we show by pharmacological and genetic approaches that Src activity was necessary to elicit optimal GDNF-mediated signaling, neurite outgrowth, and survival. In particular, p60Src, but not the other ubiquitous SFKs, Fyn and Yes, was responsible for the observed effects. Moreover, Src appeared to promote neuronal survival via a phosphatidylinositol-3 kinase (PI-3K)-dependent pathway because the PI-3K inhibitor LY294002 prevented GFL-mediated neuronal survival and prevented activated Src-mediated neuronal survival. In contrast, the inhibition of Src activity had no effects on NGF-mediated survival, indicating that the requirement for Src was selective for GFL-mediated neuronal survival. These data confirm the importance of protein-protein interactions between Ret and raft-associated proteins in the signaling pathways elicited by GDNF, and the data implicate Src as one of the major signaling molecules involved in GDNF-mediated bioactivity.


Subject(s)
Drosophila Proteins , Nerve Growth Factors/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors , Membrane Microdomains/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurites/metabolism , Neurons/cytology , Neurons/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-fyn , Proto-Oncogene Proteins c-ret , Proto-Oncogene Proteins c-yes , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/metabolism , Proto-Oncogene Proteins pp60(c-src)/pharmacology , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/pharmacology
19.
J Neurosci Methods ; 100(1-2): 63-9, 2000 Jul 31.
Article in English | MEDLINE | ID: mdl-11040367

ABSTRACT

One of the more time-consuming procedures in the study of exogenously expressed proteins in cell lines is the selection of individual transfected clones. In recent years, green fluorescent protein variants with excitation/emission spectra matching the typical flow cytometer configurations have been generated and are in common use. We employed PC12 cells transfected with vectors encoding fluorescent proteins and a fluorescence selection procedure using a fluorescence-activated cell-sorter. In order to select the optimal co-electroporation and sorting conditions, we used the simultaneous detection of two variants of the green fluorescent protein, that possess separable emission peaks when excited at 488 nm. Using these variants and the adequate combination of band-pass filters, we were able to analyze and establish the conditions for identifying and sorting cells transfected with enhanced green fluorescent protein, that simultaneously express another plasmid of interest. Using this procedure, the cells sorted that express both plasmids exceeded 90%. The whole procedure did not alter the physiological responsiveness of the transfected cells to growth factors, and has been successfully applied to the constitutive activation of the mitogen-activated protein kinase pathway, resulting in the spontaneous differentiation of PC12 cells. Also, this procedure has been used with other set of expression vectors encoding proteins that protect PC12 cells from apoptosis caused by different stimuli. The method that we present here provides an easy and fast procedure to obtain a high proportion of positively transfected populations of PC12 cells.


Subject(s)
Bacterial Proteins/genetics , Flow Cytometry/methods , Genes, Reporter/genetics , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , PC12 Cells/metabolism , Transfection/methods , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Flow Cytometry/instrumentation , Flow Cytometry/standards , Green Fluorescent Proteins , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , Neurites/ultrastructure , PC12 Cells/cytology , PC12 Cells/drug effects , Rats
20.
J Biol Chem ; 275(48): 37846-52, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10982788

ABSTRACT

Nerve growth factor (NGF) induces survival and differentiation of the neural crest-derived PC12 cell line. Caveolae are cholesterol-enriched, caveolin-containing plasma membrane microdomains involved in vesicular transport and signal transduction. Here we demonstrate the presence of caveolae in PC12 cells and their involvement in NGF signaling. Our results showed the expression of caveolin-1 by Western blot and confocal immuno-microscopy. The presence of plasma membrane caveolae was directly shown by rapid-freeze deep-etching electron microscopy. Moreover, combined deep-etching and immunogold techniques revealed the presence of the NGF receptor TrkA in the caveolae of PC12 cells. These data together with the cofractionation of Shc, Ras, caveolin, and TrkA in the caveolae fraction supported a role for these plasma membrane microdomains in NGF signaling. To approach this hypothesis, caveolae were disrupted by treatment of PC12 cells with cholesterol binding drugs. Either filipin or cyclodextrin treatment increased basal levels of MAPK phosphorylation. In contrast, pretreatment of PC12 cells with these drugs inhibited the NGF- but not the epidermal growth factor-induced MAPK phosphorylation without affecting the TrkA autophosphorylation. Taken together, our results demonstrate the presence of caveolae in PC12 cells, which contain the high affinity NGF receptor TrkA, and the specific involvement of these cholesterol-enriched plasma membrane microdomains in the propagation of the NGF-induced signal.


Subject(s)
Epidermal Growth Factor/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/pharmacology , Receptor, trkA/metabolism , Signal Transduction , Animals , Caveolin 1 , Caveolins/metabolism , Cholesterol/metabolism , Microscopy, Electron , Nerve Growth Factor/metabolism , PC12 Cells , Phosphorylation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL