Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 7(6): e2287, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27362805

ABSTRACT

We previously reported that renal clear cell carcinoma cells (RCC) express both tumor necrosis factor receptor (TNFR)-1 and -2, but that, in organ culture, a TNF mutein that only engages TNFR1, but not TNFR2, causes extensive cell death. Some RCC died by apoptosis based on detection of cleaved caspase 3 in a minority TUNEL-positive cells but the mechanism of death in the remaining cells was unexplained. Here, we underpin the mechanism of TNFR1-induced cell death in the majority of TUNEL-positive RCC cells, and show that they die by necroptosis. Malignant cells in high-grade tumors displayed threefold to four fold higher expression of both receptor-interacting protein kinase (RIPK)1 and RIPK3 compared with non-tumor kidney tubular epithelium and low-grade tumors, but expression of both enzymes was induced in lower grade tumors in organ culture in response to TNFR1 stimulation. Furthermore, TNFR1 activation induced significant MLKL(Ser358) and Drp1(Ser616) phosphorylation, physical interactions in RCC between RIPK1-RIPK3 and RIPK3-phospho-MLKL(Ser358), and coincidence of phospho-MLKL(ser358) and phospho-Drp1(Ser616) at mitochondria in TUNEL-positive RCC. A caspase inhibitor only partially reduced the extent of cell death following TNFR1 engagement in RCC cells, whereas three inhibitors, each targeting a different step in the necroptotic pathway, were much more protective. Combined inhibition of caspases and necroptosis provided additive protection, implying that different subsets of cells respond differently to TNF-α, the majority dying by necroptosis. We conclude that most high-grade RCC cells express increased amounts of RIPK1 and RIPK3 and are poised to undergo necroptosis in response to TNFR1 signaling.


Subject(s)
Apoptosis , Carcinoma, Renal Cell/pathology , Epithelial Cells/metabolism , Kidney Neoplasms/pathology , Kidney Tubules/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Acrylamides/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Nick-End Labeling , Kidney Neoplasms/genetics , Necrosis , Organ Culture Techniques , Quinazolinones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
2.
Cell Death Dis ; 1: e59, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-21364664

ABSTRACT

Puma is a potent BH3-only protein that antagonises anti-apoptotic Bcl-2 proteins, promotes Bax/Bak activation and has an essential role in multiple apoptotic models. Puma expression is normally kept very low, but can be induced by several transcription factors including p53, p73, E2F1 and FOXO3a, whereby it can induce an apoptotic response. As Puma can to bind and inactivate all anti-apoptotic members of the Bcl-2 family, its activity must be tightly controlled. We report here, for the first time, evidence that Puma is subject to post-translational control through phosphorylation. We show that Puma is phosphorylated at multiple sites, with the major site of phosphorylation being serine 10. Replacing serine 10 with alanine causes reduced Puma turnover and enhanced cell death. Interestingly, Puma turnover occurs through the proteasome, and substitution of serine 10 causes elevated Puma levels independently of macroautophagy, Bcl-2 family member binding, caspase activity and apoptotic death. We conclude, therefore, that phosphorylation of Puma at serine 10 promotes Puma turnover, represses Puma's cell death potential and promotes cell survival. Owing to the highly pro-apoptotic nature of Puma, these studies highlight an important additional regulatory step in the determination of cellular life or death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis Regulatory Proteins/genetics , HeLa Cells , Humans , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Proteins/genetics
3.
Oncogene ; 28(21): 2128-41, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19347031

ABSTRACT

The binding of Bcl-2 to Beclin-1 reduces Beclin-1's capacity to induce autophagy. Here, we have tested whether the interaction is reciprocated by loss of Bcl-2's anti-apoptotic function. We targeted Bcl-2 to mitochondria or endoplasmic reticulum (ER) and induced apoptosis using several apoptotic stimuli that initiate ER and/or mitochondrial signaling pathways (UV radiation, TNF and cycloheximide, staurosporine, thapsigargin and tunicamycin). When Beclin-1 and Bcl-2 were expressed together in HeLa cells, Beclin-1 (but not Beclin-1 lacking the Bcl-2-binding domain) followed Bcl-2 to the appropriate organelle with complete or near-complete overlap (comprising 60 and 30% of cells, respectively). The interaction between Beclin-1 and Bcl-2 was verified by immunoprecipitation, and a membrane-proximate localization of Beclin-1 was shown by immunoelectron microscopy. Apoptosis was followed by measuring changes in nuclear morphology, caspase-3 activity, poly-ADP-ribose polymerase cleavage or punctation of mRFP-Bax on mitochondria. Binding of Beclin-1 to Bcl-2 did not modify apoptosis irrespective of Bcl-2 concentration, location or apoptotic stimulus. A similar result was obtained in Atg5-/- cells that are autophagy-deficient, arguing against compensation for the loss of protection by Bcl-2 by autophagy-mediated survival induced by Beclin-1. Hence, although Beclin-1 contains a BH3-only motif typical of pro-apoptotic proteins, it is a negligible modulator of Bcl-2's anti-apoptotic function.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Beclin-1 , Cells, Cultured , Gene Expression Regulation , Humans , Intracellular Space/metabolism , Membrane Proteins/genetics , Mice , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics
5.
Cell Death Differ ; 15(3): 545-54, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18174902

ABSTRACT

Caspase inhibition can extend the survival of cells undergoing apoptosis beyond the point of mitochondrial outer membrane permeabilisation (MOMP), but this does not confer long-term protection because caspase-independent death pathways emerge. Here, we describe a novel mechanism of mitochondrial self-destruction in caspase-inhibited cells, whose hallmark is the degradation of Tim23, the essential pore-forming component of the TIM23 inner membrane translocase. We show that Tim23 degradation occurs in cycling and post-mitotic cells, it is caspase-independent but Bax/Bak dependent, and it follows cytochrome c release. The proteolytic degradation of Tim23 is induced by MOMP and is mitochondrion-autonomous, as it also occurs in isolated mitochondria undergoing permeability transition. Degradation of Tim23 is selective, as expression of several other inner membrane proteins that regulate respiratory chain function is unaffected, and is not autophagic, as it occurs similarly in autophagy-proficient and -deficient (Atg-5 knockout) cells. Depleting Tim23 with siRNA is sufficient to inhibit cell proliferation and prevent long-term survival, while expression of degradation-resistant Tim23-GFP in mitochondria delays caspase-independent cell death. Thus, mitochondrial autodigestion of Tim23 joins the array of processes contributing to caspase-independent cell death. Because mitochondrial biogenesis requires a functional protein-import machinery, preventing Tim23 degradation might, therefore, be essential for repairing damaged mitochondria in chronic degenerative diseases.


Subject(s)
Apoptosis , Caspase Inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Biological Transport , Cell Cycle , Cell Survival , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , HeLa Cells , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , RNA Interference , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
6.
Neuroreport ; 14(18): 2397-401, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14663199

ABSTRACT

We have examined how herbimycin affects the survival and neuritogenesis of avian sympathetic neurons. Herbimycin promoted sympathetic neuron survival and neuritogenesis. At higher concentrations (> or = 100 ng/ml), herbimycin still enhanced neuron survival but blocked neuritogenesis. Addition of herbimycin (10-30 ng/ml) to neurons cultured in the presence of NGF or retinal conditioned medium altered neuronal morphology, with an increase in the number of neurites. Addition of NGF during hypoxia rescued 52% of the neurons compared to 14% survival in control conditions. Herbimycin alone rescued about 50% of the neurons. In the presence of NGF and 100 ng/ml herbimycin, 81% of the neurons survived hypoxia. Our results show that herbimycin promotes survival of chick sympathetic neurons and potentiates the effects of NGF.


Subject(s)
Adrenergic Fibers/drug effects , Adrenergic Fibers/physiology , Quinones/pharmacology , Animals , Benzoquinones , Cell Death/drug effects , Cell Death/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chick Embryo , Lactams, Macrocyclic , Neurites/drug effects , Neurites/physiology , Rifabutin/analogs & derivatives
7.
Dev Neurosci ; 23(2): 153-64, 2001.
Article in English | MEDLINE | ID: mdl-11509838

ABSTRACT

The role of target interactions in the development and functional maturation of peripheral neurons was investigated using an immortalized sympathetic precursor cell line. bMAH cells underwent neuronal differentiation in response to neurotrophic factors, but maintained an immature neuronal phenotype characterized by small cell bodies and continued cell division. Co-culture with cardiac myocytes, a target of sympathetic innervation, promoted the appearance of large-diameter postmitotic bMAH neurons. Analysis of bMAH maturation in the presence and absence of co-cultured myocytes indicated that myocyte-derived factors promoted the survival of maturing bMAH neurons prior to their acquisition of nerve growth factor dependence. Myocyte interactions also promoted the functional maturation of bMAH neurons, leading to an increase in the localization of synaptic vesicle proteins into neuritic varicosities and the acquisition of sympathetic-like intrinsic electrical properties. Like primary sympathetic neurons, mature bMAH neurons formed functional connections to cardiac myocytes as measured by evoked postsynaptic responses in connected myocytes. The effects of myocyte co-culture on developing bMAH neurons could be mimicked by myocyte conditioned medium, indicating that cardiac myocytes produce soluble factors that promote the appearance of mature neurons. These experiments indicate that targets of innervation play a role in directing the development and final maturation of peripheral neurons.


Subject(s)
Myocardium/cytology , Neurons/cytology , Stem Cells/cytology , Sympathetic Nervous System/cytology , Action Potentials , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Communication/physiology , Cell Differentiation , Cell Division/drug effects , Cell Line , Cytarabine/pharmacology , Electrophysiology , Neurons/physiology , Rats
8.
J Virol ; 75(8): 3885-95, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264377

ABSTRACT

A neonatal rat dorsal root ganglion-derived neuronal culture system has been utilized to study herpes simplex virus (HSV) latency establishment, maintenance, and reactivation. We present our initial characterization of viral gene expression in neurons following infection with replication-defective HSV recombinants carrying beta-galactosidase and/or green fluorescent protein reporter genes under the control of lytic cycle- or latency-associated promoters. In this system lytic virus reporter promoter activity was detected in up to 58% of neurons 24 h after infection. Lytic cycle reporter promoters were shut down over time, and long-term survival of neurons harboring latent virus genomes was demonstrated. Latency-associated promoter-driven reporter gene expression was detected in neurons from early times postinfection and was stably maintained in up to 83% of neurons for at least 3 weeks. In latently infected cultures, silent lytic cycle promoters could be activated in up to 53% of neurons by nerve growth factor withdrawal or through inhibition of histone deacetylases by trichostatin A. We conclude that the use of recombinant viruses containing reporter genes, under the regulation of lytic and latency promoter control in neuronal cultures in which latency can be established and reactivation can be induced, is a potentially powerful system in which to study the molecular events that occur during HSV infection of neurons.


Subject(s)
Ganglia, Spinal/virology , Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Neurons/virology , Promoter Regions, Genetic/genetics , Virus Activation/genetics , Virus Latency/genetics , Animals , Animals, Newborn , Cell Line , Cell Survival , Cells, Cultured , Cytomegalovirus/genetics , DNA, Recombinant/genetics , Ganglia, Spinal/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Reporter/genetics , Herpesvirus 1, Human/drug effects , Hydroxamic Acids/pharmacology , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Virus Activation/drug effects , Virus Latency/drug effects
9.
Curr Biol ; 11(5): 361-5, 2001 Mar 06.
Article in English | MEDLINE | ID: mdl-11267874

ABSTRACT

Pan caspase inhibitors are potentially powerful cell-protective agents that block apoptosis in response to a wide variety of insults that cause tissue degeneration. In many conditions, however, the blockade of apoptosis by caspase inhibitors does not permit long-term cell survival, but the reasons are not entirely clear. Here we show that the blockade of apoptosis by Boc.Aspartyl(O-methyl)CH2F can result in the highly selective elimination of the entire cohort of mitochondria, including mitochondrial DNA, from both neurons and HeLa cells, irrespective of the stimulus used to trigger apoptosis. In cells that lose their mitochondria, the nuclear DNA, Golgi apparatus, endoplasmic reticulum, centrioles, and plasma membrane remain undamaged. The capacity to remove mitochondria is both specific and regulated since mitochondrial loss in neurons is completely prevented by the expression of the antiapoptotic protein Bcl-2 and partially suppressed by the autolysosomal inhibitor bafilomycin. Cells without mitochondria are more tolerant to an anaerobic environment but are essentially irreversibly committed to death. Prevention of mitochondrial loss may be crucial for the long-term regeneration of tissues emerging from an apoptotic episode in which death was prevented by caspase blockade.


Subject(s)
Apoptosis , Caspase Inhibitors , Mitochondria/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Eukaryotic Cells/physiology , HeLa Cells , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
10.
J Biol Chem ; 276(7): 4531-4, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11121395

ABSTRACT

Studies in non-neuronal cells show that c-Jun N-terminal kinases (JNK) play a key role in apoptotic cell death. In some neurons JNK is also thought to initiate cell death by the activation of c-Jun. JNK inhibition has been achieved pharmacologically by inhibiting upstream kinases, but there has been no direct demonstration that inhibition of JNK can prevent neuronal death. We have therefore examined whether the JNK binding domain (JBD) of JNK-interacting protein-1 (JIP-1, a scaffold protein and specific inhibitor of JNK) can inhibit c-Jun phosphorylation and support the survival of sympathetic neurons deprived of NGF. We show that expression of the JBD in >80% of neurons was sufficient to prevent the phosphorylation of c-Jun and its nuclear accumulation as well as abrogate neuronal cell death induced by NGF deprivation. JBD expression also preserved the capacity of mitochondria to reduce MTT. Interestingly, although the PTB domain of JIP was reported to interact with rhoGEF, expression of the JBD domain was sufficient to localize the protein to the membrane cortex and growth cones. Hence, JNK activation is a key event in apoptotic death induced by NGF withdrawal, where its point of action lies upstream of mitochondrial dysfunction.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/chemistry , Ganglia, Sympathetic/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Cell Culture Techniques , JNK Mitogen-Activated Protein Kinases , Nerve Growth Factor/physiology , Neurons/physiology , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-jun/metabolism , Rats , Transfection
11.
Curr Biol ; 10(18): 1151-4, 2000 Sep 21.
Article in English | MEDLINE | ID: mdl-10996800

ABSTRACT

Phosphorylation of BAD, a pro-apoptotic member of the Bcl-2 protein family, on either Ser112 or Ser136 is thought to be necessary and sufficient for growth factors to promote cell survival. Here we report that Ser155, a site phosphorylated by protein kinase A (PKA), also contributes to cell survival. Ser112 is thought to be the critical PKA target, but we found that BAD fusion proteins containing Ala at Ser112 (S112A) or Ser136 (S136A) or at both positions (S112/136A) were still heavily phosphorylated by PKA in an in vitro kinase assay. BAD became insensitive to phosphorylation by PKA only when both Ser112 and Ser136, or all three serines (S112/136/155) were mutated to alanine. In HEK293 cells, BAD fusion proteins mutated at Ser155 were refractory to phosphorylation induced by elevation of cyclic AMP(cAMP) levels. Phosphorylation of the S112/136A mutant was >90% inhibited by H89, a PKA inhibitor. The S155A mutant induced more apoptosis than the wild-type protein in serum-maintained CHO-K1 cells, and apoptosis induced by the S112/136A mutant was potentiated by serum withdrawal. These data suggest that Ser155 is a major site of phosphorylation by PKA and serum-induced kinases. Like Ser112 and Ser136, phosphorylation of Ser155 contributes to the cancellation of the pro-apoptotic function of BAD.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Phosphoserine/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Amino Acid Substitution , Animals , CHO Cells , Carrier Proteins/chemistry , Cell Line , Cell Survival , Colforsin/pharmacology , Cricetinae , Culture Media, Serum-Free , Humans , Kidney , Phosphorylation , Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , bcl-Associated Death Protein
12.
J Cell Biol ; 150(4): 741-54, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10953000

ABSTRACT

Axotomized neurons have several characteristics that are different from intact neurons. Here we show that, unlike established cultures, the axotomized sympathetic neurons deprived of NGF become committed to die before caspase activation, since the same proportion of NGF-deprived neurons are rescued by NGF regardless of whether caspases are inhibited by the pan-caspase inhibitor Boc-Asp(O-methyl)-CH(2)F (BAF). Despite prolonged Akt and ERK signaling induced by NGF after BAF treatment has prevented death, the neurons fail to increase protein synthesis, recover ATP levels, or grow. Within 3 d, all the mitochondria disappear without apparent removal of any other organelles or loss of membrane integrity. Although NGF does rescue intact BAF-treated 6-d cultures after NGF deprivation, rescue by NGF fails when these neurons are axotomized before NGF deprivation and BAF treatment. Moreover, cytosolic cytochrome c rapidly kills axotomized neurons. We propose that axotomy induces signals that make sympathetic neurons competent to die prematurely. NGF cannot repair these NGF-deprived, BAF-treated neurons because receptor signaling (which is normal) is uncoupled from protein renewal, and the mitochondria (which are damaged) go on to be eliminated. Hence, the order of steps underlying neuronal death commitment is mutable and open to regulation.


Subject(s)
Aspartic Acid/analogs & derivatives , Nerve Growth Factor/physiology , Neurons/cytology , Neurons/physiology , Protein Serine-Threonine Kinases , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Aspartic Acid/pharmacology , Axotomy , Caspase Inhibitors , Caspases/metabolism , Cell Division/drug effects , Cell Survival , Cells, Cultured , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/pharmacology , Neurites/physiology , Neurites/ultrastructure , Neurons/drug effects , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology
13.
Mol Cell Neurosci ; 16(2): 97-110, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10924254

ABSTRACT

Bcl-2 overexpression prevents neuronal death after injury or neurotrophic factor-deprivation but the biochemical consequences of survival maintenance by Bcl-2 have hardly been explored. We show that unlike NGF, adenovirally delivered hBcl-2 supports the survival of over 80% of the neurons without activating ERK and Akt phosphorylation, or suppressing JNK phosphorylation, or enhancing cell growth. However, the proapoptotic protein BAD, whose phosphorylation is induced by NGF, is degraded in NGF-deprived neurons expressing hBcl-2, while the level of Bcl-xL remains unaffected. Interestingly, degradation of BAD protein is prevented by the pan-caspase inhibitor Boc.Asp(OMe)fmk. We propose that NGF-deprivation promotes dephosphorylation of BAD while hBcl-2 facilitates its release into the cytoplasm where it is degraded by noncaspase, Boc.Asp(O-Me)fmk-inhibitable proteases. The potential importance of BAD degradation is suggested by our finding that overexpressed BAD kills NGF-maintained sympathetic neurons by apoptosis, while hBcl-2 prevents BAD-induced death.


Subject(s)
Carrier Proteins/metabolism , Nerve Growth Factor/pharmacology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Sympathetic Nervous System/cytology , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Gene Expression/physiology , Genetic Vectors , Neurons/cytology , Phosphorylation , Rats , Serine/metabolism , Signal Transduction/genetics , bcl-Associated Death Protein
14.
J Biol Chem ; 275(12): 8817-24, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722727

ABSTRACT

Ras promotes robust survival of many cell systems by activating the phosphatidylinositol 3-kinase (PI3-kinase)/Akt pathway, but little is understood about the survival functions of the Ras/ERK pathway. We have used three different effector-loop mutant forms of Ras, each of which activates a single downstream effector pathway, to dissect their individual contributions to survival of nerve growth factor (NGF)-dependent sympathetic neurons. The PI3-kinase pathway-selective protein Ras(Val-12)Y40C was as powerful as oncogenic Ras(Val-12) in preventing apoptosis induced by NGF deprivation but conferred no protection against apoptosis induced by cytosine arabinoside. Identical results were obtained with transfected Akt. In contrast, the ERK pathway-selective protein Ras(Val-12)T35S had no protective effects on NGF-deprived neurons but was almost as strongly protective as Ras(Val-12) against cytosine arabinoside-induced apoptosis. The protective effects of Ras(Val-12)T35S against cytosine arabinoside were completely abolished by the ERK pathway inhibitor PD98059. Ras(Val-12)E37G, an activator of RalGDS, had no survival effect on either death pathway, similar to RasS17N, the full survival antagonist. Thus, Ras provides two independent survival pathways each of which inhibits a distinct apoptotic mechanism. Our study presents one of the few clear-cut cases where only the Ras/ERK, but not the Ras/PI3K/Akt pathway, plays a dominant survival signaling role.


Subject(s)
Apoptosis , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins p21(ras)/metabolism , Superior Cervical Ganglion/metabolism , Animals , Cell Survival , Cells, Cultured , Cytarabine/pharmacology , Models, Biological , Nerve Growth Factor/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Recombinant Proteins/metabolism , Signal Transduction , Superior Cervical Ganglion/cytology , ral Guanine Nucleotide Exchange Factor/metabolism
15.
Mol Cell Neurosci ; 14(3): 180-98, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10576889

ABSTRACT

Autophagy is a mechanism whereby cells digest themselves from within and so may be used in lieu of apoptosis to execute cell death. Little is known about its role in neurons. In newly isolated sympathetic neurons, two independent apoptotic stimuli, NGF-deprivation or cytosine arabinoside added in the presence of NGF, caused a 30-fold increase in autophagic particle numbers, many autophagosomes appearing before any signs of DNA-fragmentation. The anti-autophagic drug 3-methyladenine also delayed apoptosis, its neuroprotection correlating with inhibition of cytochrome c release from mitochondria and prevention of caspase activation. In contrast, autophagic activity remained elevated in neurons treated with the pan-caspase inhibitor Boc-Asp(OMe)fmk, which inhibited morphological apoptosis but did not inhibit cytochrome c release nor prevent cell death. We propose that the same apoptotic signals that cause mitochondrial dysfunction also activate autophagy. Once activated, autophagy may mediate caspase-independent neuronal death.


Subject(s)
Apoptosis , Neurons/cytology , Neurons/physiology , Signal Transduction/physiology , Superior Cervical Ganglion/cytology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Autophagy/drug effects , Autophagy/physiology , Caspase Inhibitors , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Mitochondria/drug effects , Mitochondria/ultrastructure , Neurons/ultrastructure , Rats , Rats, Wistar , Superior Cervical Ganglion/physiology
16.
J Neurochem ; 73(5): 1901-12, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537048

ABSTRACT

The c-Jun N-terminal kinase signaling cascade appears to play a role in some cases of cell death, including neuronal apoptosis. CEP-1347 (KT7515), an indolocarbazole of the K252a family, blocks this stress signaling cascade and promotes survival. Here, we used CEP-1347 to probe whether neuronal death pathways activated by distinct insults also possess elements in common. Cultured rat sympathetic neurons and neuronally differentiated PC12 cells were induced to die by withdrawal of nerve growth factor, exposure to ultraviolet irradiation, or subjection to oxidative stress. In each case, death was prevented by 100-200 nM CEP-1347. Moreover, in each of these death paradigms, c-Jun N-terminal kinase 1 activity in neuronally differentiated PC12 cells was elevated by two- or threefold, and this increase was totally blocked by CEP-1347 at concentrations that promoted survival. In contrast, 200 nM CEP-1347 did not block death due to serum withdrawal from undifferentiated PC12 cells or to activation of Fas in Jurkat T cell cultures, even though in each case c-Jun N-terminal kinase 1 activation occurred and was inhibited by CEP-1347. These observations suggest that some but not all death pathways triggered by different insults can include a common mechanistic component, a likely candidate for which is activation of the c-Jun N-terminal kinase signaling cascade.


Subject(s)
Carbazoles/pharmacology , Cell Death/drug effects , Enzyme Inhibitors/pharmacology , Ganglia, Sympathetic/cytology , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurons/physiology , Animals , Cell Differentiation , Cells, Cultured , Enzyme Activation/drug effects , Humans , Jurkat Cells , MAP Kinase Kinase 4 , Neurites/physiology , Oxidative Stress , PC12 Cells , Rats , Rats, Sprague-Dawley , Signal Transduction , fas Receptor/physiology
17.
Brain Res ; 837(1-2): 127-42, 1999 Aug 07.
Article in English | MEDLINE | ID: mdl-10433995

ABSTRACT

Phosphoinositide 3-kinase and its downstream effector kinase PKB/Akt have been suggested to have crucial roles in suppressing apoptosis in several classes of neurons. However, few studies have conducted a long-term investigation of either kinase activity, many studies relying instead on use of the phosphoinositide 3-kinase inhibitors wortmannin and LY294002. When we added LY294002 or wortmannin to sympathetic neurons, apoptosis in the presence of nerve growth factor (NGF) was very slow compared to that obtained by NGF deprivation. However, expression of a kinase-inactive mutant of PKB/Akt in the presence of NGF induced apoptosis in a significant proportion of the neurons. To understand this discrepancy, we investigated more closely the regulation of PKB/Akt activity by NGF. NGF stimulation induced a rapid increase in PKB/Akt activity which was sustained at approximately 6-fold up to 24 h. Phosphoinositide 3-kinase was also rapidly activated by NGF. However, concentrations of wortmannin which completely blocked phosphoinositide 3-kinase activity in the neurons inhibited no more than 50-70% of cellular PKB/Akt activity. Similarly, approximately 50% of maximal NGF-stimulated PKB/Akt activity remained elevated at concentrations of LY294002 which completely blocked neurite outgrowth, a process known to be phosphoinositide 3-kinase dependent. We suggest that a proportion of the sustained PKB/Akt activity induced by NGF is mediated by phosphoinositide 3-kinase-independent pathways. These results raise a cautionary note as to the usefulness of LY294002 or wortmannin as tools to dissect the role of PKB/Akt in neuronal survival.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Superior Cervical Ganglion/physiology , Androstadienes/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Kinetics , Morpholines/pharmacology , Neurites/drug effects , Neurons/cytology , Neurons/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/metabolism , Superior Cervical Ganglion/cytology , Transfection , Wortmannin
18.
J Neurosci ; 19(2): 664-73, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880587

ABSTRACT

The antimitotic nucleoside cytosine arabinoside (araC) causes apoptosis in postmitotic neurons for which two mechanisms have been suggested: (1) araC directly inhibits a trophic factor-maintained signaling pathway required for survival, effectively mimicking trophic factor withdrawal; and (2) araC induces apoptosis by a p53-dependent mechanism distinct from trophic factor withdrawal. In rat sympathetic neurons, we found that araC treatment for 12 hr induced approximately 25% apoptosis without affecting NGF-maintained signaling; there was neither reduction in the activity of mitogen activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) or protein kinase B/Akt, a kinase implicated in NGF-mediated survival, nor was there c-Jun N-terminal kinase (JNK) activation or c-Jun N-terminal phosphorylation, events implicated in apoptosis induced by NGF withdrawal. However, araC treatment, but not NGF-withdrawal, elevated expression of p53 protein before and during apoptosis. Additionally, araC-induced apoptosis was suppressed in sympathetic neurons from p53 null mice. Although MAPK/ERK activity is not necessary for NGF-induced survival, it protected against toxicity by araC, because inhibition of the MAPK pathway by PD98059 resulted in a significant increase in the rate of apoptosis induced by araC in the presence of NGF. Consistent with this finding, ciliary neurotrophic factor, which does not cause sustained activation of MAPK/ERK, did not protect against araC toxicity. Our data show that, in contrast to NGF deprivation, araC induces apoptosis via a p53-dependent, JNK-independent mechanism, against which MAPK/ERK plays a substantial protective role. Thus, NGF can suppress apoptotic mechanisms in addition to those caused by its own deprivation.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytarabine/pharmacology , Genes, p53/physiology , Neurons/drug effects , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-jun/physiology , Sympathetic Nervous System/drug effects , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Mice , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Neurons/enzymology , Neurons/physiology , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sympathetic Nervous System/cytology , Sympathetic Nervous System/enzymology
19.
Genes Dev ; 12(4): 557-70, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9472024

ABSTRACT

The activation of MAPKs is controlled by the balance between MAPK kinase and MAPK phosphatase activities. The latter is mediated by a subset of phosphatases with dual specificity (VH-1 family). Here, we describe a new member of this family encoded by the puckered gene of Drosophila. Mutations in this gene lead to cytoskeletal defects that result in a failure in dorsal closure related to those associated with mutations in basket, the Drosophila JNK homolog. We show that puckered mutations result in the hyperactivation of DJNK, and that overexpression of puc mimics basket mutant phenotypes. We also show that puckered expression is itself a consequence of the activity of the JNK pathway and that during dorsal closure, JNK signaling has a dual role: to activate an effector, encoded by decapentaplegic, and an element of negative feedback regulation encoded by puckered.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drosophila Proteins , Drosophila/embryology , Gene Expression Regulation, Enzymologic , Insect Proteins/genetics , Mitogen-Activated Protein Kinases , Phosphoprotein Phosphatases/genetics , Amino Acid Sequence , Animals , Base Sequence , Drosophila/enzymology , Drosophila/genetics , Feedback , Genes, Insect , Insect Proteins/metabolism , JNK Mitogen-Activated Protein Kinases , Models, Biological , Molecular Sequence Data , Morphogenesis/genetics , Phosphoprotein Phosphatases/metabolism , Sequence Homology, Amino Acid , Signal Transduction
20.
J Neurochem ; 69(2): 550-61, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9231712

ABSTRACT

We have investigated the relationship between c-Jun N-terminal kinase (JNK) activity, apoptosis, and the potential of survival factors to rescue primary rat sympathetic neurones deprived of trophic support. Incubation of sympathetic neurones in the absence of nerve growth factor (NGF) caused a time-dependent increase in JNK activity, which became apparent by 3 h and attained maximal levels that were three- to fourfold higher than activity measured in neurones maintained for the same periods with NGF. Continuous culture in the presence of either NGF or the cyclic AMP analogue 4-(8-chlorophenylthio) cyclic AMP (CPTcAMP) not only prevented JNK activation from occurring, but also suppressed JNK activity that had been elevated by prior culture of the neurones in the absence of trophic support. When either NGF or CPTcAMP was added to cultures that had been initially deprived of neurotrophic support for up to 10 h, this resulted in complete suppression of total JNK activity, arrest of apoptosis, and rescue of >90% of the neurones that did not display apoptotic morphology by this time. However, when either agent was added after more protracted periods of initial neurotrophin deprivation (> or = 14 h), although this also resulted in near-complete suppression of total JNK activity and short-term arrest of apoptosis, not all of the neurones that appeared to be nonapoptotic at the time of agent addition were rescued. The lack of death commitment after 10 h of maintained JNK activity was not due to a late induction of c-Jun expression, because the majority of newly isolated sympathetic neurones had already been expressing high levels of c-Jun in their nuclei for several hours, yet were capable of being rescued by NGF. Elevation of JNK activity as a result of neurotrophic-factor deprivation was also associated with enhanced phosphorylation of c-Jun, assessed by immunoblot analysis and immunocytochemistry, and addition of NGF to cultures previously deprived of neurotrophic support resulted in a reversion of the state of phospho-c-Jun to that observed in cultures that had been maintained in the continuous presence of trophic support. We conclude that activation of JNK and c-Jun phosphorylation are not necessarily rate-limiting for apoptosis induction. In some neurones undergoing prolonged NGF deprivation, suppression of JNK activity and c-Jun dephosphorylation by NGF may be insufficient to effect their rescue. Thus, if c-Jun mediates death by increasing the expression of "death" genes, these must become effective very close to the death commitment point.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins c-jun/metabolism , Superior Cervical Ganglion/metabolism , Animals , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Enzyme Activation/drug effects , Immunoblotting , JNK Mitogen-Activated Protein Kinases , Kinetics , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/pharmacology , Phosphorylation , Rats , Superior Cervical Ganglion/cytology , Thionucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...