Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
2.
Neurobiol Dis ; 136: 104743, 2020 03.
Article in English | MEDLINE | ID: mdl-31931138

ABSTRACT

Rho GTPases play a central role in neuronal survival; however, the antagonistic relationship between Rac and Rho in the regulation of motor neuron survival remains poorly defined. In the current study, we demonstrate that treatment with NSC23766, a selective inhibitor of the Rac-specific guanine nucleotide exchange factors, Tiam1 and Trio, is sufficient to induce the death of embryonic stem cell (ESC)-derived motor neurons. The mode of cell death is primarily apoptotic and is characterized by caspase-3 activation, de-phosphorylation of ERK5 and AKT, and nuclear translocation of the BH3-only protein Bad. As opposed to the inhibition of Rac, motor neuron cell death is also induced by constitutive activation of Rho, via a mechanism that depends on Rho kinase (ROCK) activity. Investigation of Rac and Rho in the G93A mutant, human Cu, Zn-superoxide dismutase (hSOD1) mouse model of amyotrophic lateral sclerosis (ALS), revealed that active Rac1-GTP is markedly decreased in spinal cord motor neurons of transgenic mice at disease onset and end-stage, when compared to age-matched wild type (WT) littermates. Furthermore, although there is no significant change in active RhoA-GTP, total RhoB displays a striking redistribution from motor neuron nuclei in WT mouse spinal cord to motor neuron axons in end-stage G93A mutant hSOD1 mice. Collectively, these data suggest that the intricate balance between pro-survival Rac signaling and pro-apoptotic Rho/ROCK signaling is critical for motor neuron survival and therefore, disruption in the balance of their activities and/or localization may contribute to the death of motor neurons in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , GTP Phosphohydrolases/metabolism , Motor Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Superoxide Dismutase/physiology , rho-Associated Kinases/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Death/physiology , Female , GTP Phosphohydrolases/genetics , Male , Mice , Mice, Transgenic , Motor Neurons/pathology , Mutation/physiology , Proto-Oncogene Proteins c-akt/genetics , rho-Associated Kinases/genetics
3.
Front Cell Neurosci ; 12: 15, 2018.
Article in English | MEDLINE | ID: mdl-29440992

ABSTRACT

The molecular mechanisms leading to motor neuron death in amyotrophic lateral sclerosis (ALS) are unknown; however, several studies have provided evidence of a central role for intrinsic apoptosis. Bcl-2 homology-3 domain (BH3)-only proteins are pro-apoptotic members of the Bcl-2 family whose enhanced expression acts as a trigger for the intrinsic apoptotic cascade. Here, we compared the relative expression of BH3-only proteins in the spinal cord of end-stage G93A mutant SOD1 mice to age-matched wild-type (WT) mice. Large alpha motor neurons in lumbar spinal cord sections of both WT and end-stage mutant SOD1 mice stained positively for a number of BH3-only proteins; however, no discernible differences were observed in either the relative intensity of staining or number of BH3-immunoreactive motor neurons between WT and mutant SOD1 mice. On the other hand, we observed significantly enhanced staining for Bid, DP5/Hrk, and BNip3L in GFAP-positive astrocytes only in end-stage G93A mutant SOD1 spinal cord. Staining of additional end-stage G93A mutant SOD1 tissues showed specific upregulation of DP5/Hrk in lumbar spinal cord sections, but not in cerebellum or cortex. Finally, examination of protein expression using western blotting also revealed marked increases in DP5/Hrk and BNip3L in G93A mutant SOD1 lumbar spinal cord lysates compared to WT controls. The upregulation of a specific subset of BH3-only proteins, including Bid, DP5/Hrk, and BNip3L, in reactive astrocytes suggests that these proteins may execute a novel non-apoptotic function within astrocytes to promote ALS disease progression, thus providing a new potential target for therapeutic intervention.

4.
J Biol Chem ; 290(15): 9363-76, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25666619

ABSTRACT

Rho family GTPases play integral roles in neuronal differentiation and survival. We have shown previously that Clostridium difficile toxin B (ToxB), an inhibitor of RhoA, Rac1, and Cdc42, induces apoptosis of cerebellar granule neurons (CGNs). In this study, we compared the effects of ToxB to a selective inhibitor of the Rac-specific guanine nucleotide exchange factors Tiam1 and Trio (NSC23766). In a manner similar to ToxB, selective inhibition of Rac induces CGN apoptosis associated with enhanced caspase-3 activation and reduced phosphorylation of the Rac effector p21-activated kinase. In contrast to ToxB, caspase inhibitors do not protect CGNs from targeted inhibition of Rac. Also dissimilar to ToxB, selective inhibition of Rac does not inhibit MEK1/2/ERK1/2 or activate JNK/c-Jun. Instead, targeted inhibition of Rac suppresses distinct MEK5/ERK5, p90Rsk, and Akt-dependent signaling cascades known to regulate the localization and expression of the Bcl-2 homology 3 domain-only protein Bad. Adenoviral expression of a constitutively active mutant of MEK5 is sufficient to attenuate neuronal cell death induced by selective inhibition of Rac with NSC23766 but not apoptosis induced by global inhibition of Rho GTPases with ToxB. Collectively, these data demonstrate that global suppression of Rho family GTPases with ToxB causes a loss of MEK1/2/ERK1/2 signaling and activation of JNK/c-Jun, resulting in diminished degradation and enhanced transcription of the Bcl-2 homology 3 domain-only protein Bim. In contrast, selective inhibition of Rac induces CGN apoptosis by repressing unique MEK5/ERK5, p90Rsk, and Akt-dependent prosurvival pathways, ultimately leading to enhanced expression, dephosphorylation, and mitochondrial localization of proapoptotic Bad.


Subject(s)
Apoptosis/physiology , MAP Kinase Signaling System/physiology , Neurons/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Aminoquinolines/pharmacology , Animals , Apoptosis/drug effects , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Cells, Cultured , Cerebellum/cytology , Female , Immunoblotting , MAP Kinase Kinase 5/metabolism , MAP Kinase Signaling System/drug effects , Male , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 7/metabolism , Neurons/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , bcl-Associated Death Protein/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/antagonists & inhibitors
5.
Mol Cell Neurosci ; 56: 322-332, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23859824

ABSTRACT

C-terminal binding proteins (CtBPs) are transcriptional co-repressors that are subject to proteasome-dependent downregulation during apoptosis. Alternative mechanisms that regulate CtBP expression are currently under investigation and the role of CtBPs in neuronal survival is largely unexplored. Here, we show that CtBPs are downregulated in cerebellar granule neurons (CGNs) induced to undergo apoptosis by a variety of stressors. Moreover, antisense-mediated downregulation of CtBP1 is sufficient to cause CGN apoptosis. Similarly, the CtBP inhibitor, 4-methylthio-2-oxobutyric acid, induces expression of the CtBP target Noxa and causes actinomycin-sensitive CGN apoptosis. Unexpectedly, we found that the mechanism of CtBP downregulation in CGNs undergoing apoptosis varies in a stimulus-specific manner involving either the proteasome or caspases. In the case of CGNs deprived of depolarizing potassium (5K apoptotic condition), caspases appear to play a dominant role in CtBP downregulation. However, incubation in 5K does not enhance the kinetics of CtBP1 degradation and recombinant CtBP1 is not cleaved in vitro by caspase-3. In addition, 5K has no significant effect on CtBP transcript expression. Finally, mouse embryonic stem cells display caspase-dependent downregulation of CtBP1 following exposure to staurosporine, an effect that is not observed in DGCR8 knockout cells which are deficient in miRNA processing. These data identify caspase-dependent downregulation of CtBPs as an alternative mechanism to the proteasome for regulation of these transcriptional co-repressors in neurons undergoing apoptosis. Moreover, caspases appear to regulate CtBP expression indirectly, at a post-transcriptional level, and via a mechanism that is dependent upon miRNA processing. We conclude that CtBPs are essential pro-survival proteins in neurons and their downregulation contributes significantly to neuronal apoptosis via the de-repression of pro-apoptotic genes.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Down-Regulation , Neurons/metabolism , Transcription Factors/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Caspase 3/metabolism , Female , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Neurons/drug effects , Neurons/physiology , Potassium/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
6.
Brain Res ; 1494: 28-43, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23220553

ABSTRACT

Neuronal cell death via apoptosis or necrosis underlies several devastating neurodegenerative diseases associated with aging. Mitochondrial dysfunction resulting from oxidative or nitrosative stress often acts as an initiating stimulus for intrinsic apoptosis or necrosis. These events frequently occur in conjunction with imbalances in the mitochondrial fission and fusion equilibrium, although the cause and effect relationships remain elusive. Here, we demonstrate in primary rat cerebellar granule neurons (CGNs) that oxidative or nitrosative stress induces an N-terminal cleavage of optic atrophy-1 (OPA1), a dynamin-like GTPase that regulates mitochondrial fusion and maintenance of cristae architecture. This cleavage event is indistinguishable from the N-terminal cleavage of OPA1 observed in CGNs undergoing caspase-mediated apoptosis (Loucks et al., 2009) and results in removal of a key lysine residue (K301) within the GTPase domain. OPA1 cleavage in CGNs occurs coincident with extensive mitochondrial fragmentation, disruption of the microtubule network, and cell death. In contrast to OPA1 cleavage induced in CGNs by removing depolarizing extracellular potassium (5K apoptotic conditions), oxidative or nitrosative stress-induced OPA1 cleavage caused by complex I inhibition or nitric oxide, respectively, is caspase-independent. N-terminal cleavage of OPA1 is also observed in vivo in aged rat and mouse midbrain and hippocampal tissues. We conclude that N-terminal cleavage and subsequent inactivation of OPA1 may be a contributing factor in the neuronal cell death processes underlying neurodegenerative diseases, particularly those associated with aging. Furthermore, these data suggest that OPA1 cleavage is a likely convergence point for mitochondrial dysfunction and imbalances in mitochondrial fission and fusion induced by oxidative or nitrosative stress.


Subject(s)
Aging/physiology , Cell Death/physiology , GTP Phosphohydrolases/metabolism , Mitochondrial Dynamics/physiology , Neurons/enzymology , Animals , Caspases/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/enzymology , Female , Hippocampus/cytology , Hippocampus/enzymology , MAP Kinase Signaling System/physiology , Male , Mesencephalon/cytology , Mesencephalon/enzymology , Mice , Nitric Oxide/metabolism , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
7.
J Biol Chem ; 287(20): 16835-48, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22378792

ABSTRACT

In several neuronal cell types, the small GTPase Rac is essential for survival. We have shown previously that the Rho family GTPase inhibitor Clostridium difficile toxin B (ToxB) induces apoptosis in primary rat cerebellar granule neurons (CGNs) principally via inhibition of Rac GTPase function. In the present study, incubation with ToxB activated a proapoptotic Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, and a pan-JAK inhibitor protected CGNs from Rac inhibition. STAT1 expression was induced by ToxB; however, CGNs from STAT1 knock-out mice succumbed to ToxB-induced apoptosis as readily as wild-type CGNs. STAT3 displayed enhanced tyrosine phosphorylation following treatment with ToxB, and a reputed inhibitor of STAT3, cucurbitacin (JSI-124), reduced CGN apoptosis. Unexpectedly, JSI-124 failed to block STAT3 phosphorylation, and CGNs were not protected from ToxB by other known STAT3 inhibitors. In contrast, STAT5A tyrosine phosphorylation induced by ToxB was suppressed by JSI-124. In addition, roscovitine similarly inhibited STAT5A phosphorylation and protected CGNs from ToxB-induced apoptosis. Consistent with these results, adenoviral infection with a dominant negative STAT5 mutant, but not wild-type STAT5, significantly decreased ToxB-induced apoptosis of CGNs. Finally, chromatin immunoprecipitation with a STAT5 antibody revealed increased STAT5 binding to the promoter region of prosurvival Bcl-xL. STAT5 was recruited to the Bcl-xL promoter region in a ToxB-dependent manner, and this DNA binding preceded Bcl-xL down-regulation, suggesting transcriptional repression. These data indicate that a novel JAK/STAT5 proapoptotic pathway significantly contributes to neuronal apoptosis induced by the inhibition of Rac GTPase.


Subject(s)
Apoptosis/drug effects , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Cerebellum/metabolism , Neurons/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , rac GTP-Binding Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Cerebellum/cytology , Mice , Mice, Knockout , Mutation , Neurons/cytology , Phosphorylation/drug effects , Rats , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , Signal Transduction/genetics , Triterpenes/pharmacology , bcl-X Protein/genetics , bcl-X Protein/metabolism
8.
Neurotox Res ; 19(3): 374-88, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20333497

ABSTRACT

1-Methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity has previously been attributed to either caspase-dependent apoptosis or caspase-independent cell death. In the current study, we found that MPP(+) induces a unique, non-apoptotic nuclear morphology coupled with a caspase-independent but calpain-dependent mechanism of cell death in primary cultures of rat cerebellar granule neurons (CGNs). Using a terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) assay in CGNs exposed to MPP(+), we observed that these neurons are essentially devoid of caspase-dependent DNA fragments indicative of apoptosis. Moreover, proteolysis of a well recognized caspase-3 substrate, poly (ADP ribose) polymerase (PARP), was not observed in CGNs exposed to MPP(+). In contrast, calpain-dependent proteolysis of fodrin and pro-caspases-9 and -3 occurred in this model coupled with inhibition of caspase-3/-7 activities. Notably, several key members of the Bcl-2 protein family appear to be prominent calpain targets in MPP(+)-treated CGNs. Bid and Bax were proteolyzed to truncated forms thought to have greater pro-death activity at mitochondria. Moreover, the pro-survival Bcl-2 protein was degraded to a form predicted to be inactive at mitochondria. Cyclin E was also cleaved by calpain to an active low MW fragment capable of facilitating cell cycle re-entry. Finally, MPP(+)-induced neurotoxicity in CGNs was significantly attenuated by a cocktail of calpain and caspase inhibitors in combination with the antioxidant glutathione. Collectively, these results demonstrate that caspases do not play a central role in CGN toxicity induced by exposure to MPP(+), whereas calpain cleavage of key protein targets, coupled with oxidative stress, plays a critical role in MPP(+)-induced neurotoxicity. Our findings underscore the complexity of MPP(+)-induced neurotoxicity and suggest that calpain may play a fundamental role in causing neuronal death downstream of mitochondrial oxidative stress and dysfunction.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Calpain/physiology , Cerebellum/drug effects , Cerebellum/enzymology , Neurons/drug effects , Neurons/enzymology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cells, Cultured , Cerebellum/pathology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/pathology , Humans , Molecular Sequence Data , Neurons/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
9.
Antioxid Redox Signal ; 11(3): 469-80, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18754708

ABSTRACT

Epigallocatechin-3-gallate (EGCG) is a major flavonoid component of green tea that displays antiapoptotic effects in numerous models of neurotoxicity. Although the intrinsic free radical scavenging activity of EGCG likely contributes to its antiapoptotic effect, other modes of action have also been suggested. We systematically analyzed the antiapoptotic action of EGCG in primary cultures of rat cerebellar granule neurons (CGNs). The dose-dependent protective effects of EGCG were determined after coincubation with eight different stimuli that each induced neuronal apoptosis by distinct mechanisms. Under these conditions, EGCG provided significant neuroprotection only from insults that induce apoptosis by causing mitochondrial oxidative stress. Despite this selective antiapoptotic effect, EGCG did not significantly alter the endogenous activities or expression of Mn(2+)- superoxide dismutase, glutathione peroxidase, Nrf2, or Bcl-2. Subfractionation of CGNs after incubation with (3)H-EGCG revealed that a striking 90-95% of the polyphenol accumulated in the mitochondrial fraction. These data demonstrate that EGCG selectively protects neurons from apoptosis induced by mitochondrial oxidative stress. This effect is likely due to accumulation of EGCG in the mitochondria, where it acts locally as a free radical scavenger. These properties of EGCG make it an interesting therapeutic candidate for neurodegenerative diseases involving neuronal apoptosis triggered by mitochondrial oxidative stress.


Subject(s)
Apoptosis/drug effects , Catechin/analogs & derivatives , Mitochondria/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Tea/chemistry , Animals , Catechin/metabolism , Catechin/pharmacology , Immunohistochemistry , Mitochondria/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley
10.
Brain Res ; 1250: 63-74, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19046944

ABSTRACT

The critical processes of mitochondrial fission and fusion are regulated by members of the dynamin family of GTPases. Imbalances in mitochondrial fission and fusion contribute to neuronal cell death. For example, increased fission mediated by the dynamin-related GTPase, Drp1, or decreased fusion resulting from inactivating mutations in the OPA1 GTPase, causes neuronal apoptosis and/or neurodegeneration. Recent studies indicate that post-translational processing regulates OPA1 function in non-neuronal cells and moreover, aberrant processing of OPA1 is induced during apoptosis. To date, the post-translational processing of OPA1 during neuronal apoptosis has not been examined. Here, we show that cerebellar granule neurons (CGNs) or neuroblastoma cells exposed to pro-apoptotic stressors display a novel N-terminal cleavage of OPA1 which is blocked by either pan-caspase or caspase-8 selective inhibitors. OPA1 cleavage occurs concurrently with mitochondrial fragmentation and cytochrome c release in CGNs deprived of depolarizing potassium (5K condition). Although a caspase-8 selective inhibitor prevents both 5K-induced OPA1 cleavage and mitochondrial fragmentation, recombinant caspase-8 fails to cleave OPA1 in vitro. In marked contrast, either caspase-8 or caspase-3 stimulates OPA1 cleavage in digitonin-permeabilized rat brain mitochondria, suggesting that OPA1 is cleaved by an intermembrane space protease which is regulated by active caspases. Finally, the N-terminal truncation of OPA1 induced during neuronal apoptosis removes an essential residue (K301) within the GTPase domain. These data are the first to demonstrate OPA1 cleavage during neuronal apoptosis and they implicate caspases as indirect regulators of OPA1 processing in degenerating neurons.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , GTP Phosphohydrolases/metabolism , Neurons/cytology , Neurons/enzymology , Animals , Brain/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase Inhibitors , Cells, Cultured , Cerebellum/enzymology , Cytochromes c/metabolism , Digitonin/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/enzymology , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology
11.
J Biol Chem ; 282(40): 29296-304, 2007 Oct 05.
Article in English | MEDLINE | ID: mdl-17690097

ABSTRACT

Bcl-2 protects cells against mitochondrial oxidative stress and subsequent apoptosis. However, the mechanism underlying the antioxidant function of Bcl-2 is currently unknown. Recently, Bax and several Bcl-2 homology-3 domain (BH3)-only proteins (Bid, Puma, and Noxa) have been shown to induce a pro-oxidant state at mitochondria (1-4). Given the opposing effects of Bcl-2 and Bax/BH3-only proteins on the redox state of mitochondria, we hypothesized that the antioxidant function of Bcl-2 is antagonized by its interaction with the BH3 domains of pro-apoptotic family members. Here, we show that BH3 mimetics that bind to a hydrophobic surface (the BH3 groove) of Bcl-2 induce GSH-sensitive mitochondrial dysfunction and apoptosis in cerebellar granule neurons. BH3 mimetics displace a discrete mitochondrial GSH pool in neurons and suppress GSH transport into isolated rat brain mitochondria. Moreover, BH3 mimetics and the BH3-only protein, Bim, inhibit a novel interaction between Bcl-2 and GSH in vitro. These results suggest that Bcl-2 regulates an essential pool of mitochondrial GSH and that this regulation may depend upon Bcl-2 directly interacting with GSH via the BH3 groove. We conclude that this novel GSH binding property of Bcl-2 likely plays a central role in its antioxidant function at mitochondria.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/chemistry , Glutathione/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Brain/metabolism , Mitochondria/metabolism , Neurons/metabolism , Oxidants/metabolism , Oxidative Stress , Protein Binding , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
12.
J Neurochem ; 94(4): 1025-39, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16092944

ABSTRACT

Rho GTPases are key transducers of integrin/extracellular matrix and growth factor signaling. Although integrin-mediated adhesion and trophic support suppress neuronal apoptosis, the role of Rho GTPases in neuronal survival is unclear. Here, we have identified Rac as a critical pro-survival GTPase in cerebellar granule neurons (CGNs) and elucidated a death pathway triggered by its inactivation. GTP-loading of Rac1 was maintained in CGNs by integrin-mediated (RGD-dependent) cell attachment and trophic support. Clostridium difficile toxin B (ToxB), a specific Rho family inhibitor, induced a selective caspase-mediated degradation of Rac1 without affecting RhoA or Cdc42 protein levels. Both ToxB and dominant-negative N17Rac1 elicited CGN apoptosis, characterized by cytochrome c release and activation of caspase-9 and -3, whereas dominant-negative N19RhoA or N17Cdc42 did not cause significant cell death. ToxB stimulated mitochondrial translocation and conformational activation of Bax, c-Jun activation, and induction of the BH3-only protein Bim. Similarly, c-Jun activation and Bim induction were observed with N17Rac1. A c-jun N-terminal protein kinase (JNK)/p38 inhibitor, SB203580, and a JNK-specific inhibitor, SP600125, significantly decreased ToxB-induced Bim expression and blunted each subsequent step of the apoptotic cascade. These results indicate that Rac acts downstream of integrins and growth factors to promote neuronal survival by repressing c-Jun/Bim-mediated mitochondrial apoptosis.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Cerebellum/physiology , Membrane Proteins/physiology , Mitochondria/physiology , Proto-Oncogene Proteins c-jun/physiology , Proto-Oncogene Proteins/physiology , rac1 GTP-Binding Protein/physiology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Bcl-2-Like Protein 11 , Caspases/metabolism , Cell Adhesion/physiology , Cell Survival/physiology , Cells, Cultured , Cytochromes c/metabolism , Enzyme Activation/drug effects , Genes, Dominant , Integrins/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Neurons/physiology , Rats , Rats, Sprague-Dawley , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors
13.
J Neurochem ; 94(1): 22-36, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15953346

ABSTRACT

Primary cerebellar granule neurons (CGNs) require depolarizing extracellular potassium for their survival. Removal of depolarizing potassium triggers CGN apoptosis that requires induction of Bim, a BH3-only Bcl-2 family member. Bim is classically thought to promote apoptosis by neutralizing pro-survival Bcl-2 proteins. To determine if this is the principal function of Bim in CGNs, we contrasted Bim-mediated apoptosis to neuronal death induced by HA14-1, a BH3-domain mimetic that antagonizes Bcl-2 and Bcl-x(L). HA14-1 elicited CGN apoptosis characterized by caspase 3 and 9 activation, cytochrome c release, conformational activation of Bax, and mitochondrial depolarization. HA14-1 provoked CGN apoptosis in the absence of Bim induction and negative regulators of Bim transcription did not prevent HA14-1-induced cell death. However, the antioxidant glutathione and its precursor, N-acetyl-l-cysteine, suppressed HA14-1-induced apoptosis. Similarly, apoptosis induced by either a structurally distinct Bcl-2/Bcl-x(L) inhibitor (compound 6) or Bcl-2 antisense oligonucleotides was diminished by glutathione. In contrast, antioxidants had no effect on CGN apoptosis provoked by either removal of depolarizing potassium or overexpression of a GFP-Bim fusion protein, two models of Bim-dependent death. These data show that antagonism of Bcl-2/Bcl-x(L) function elicits oxidative stress-dependent CGN apoptosis that is mechanistically distinct from Bim-mediated cell death. These results further indicate that, although Bcl-2/Bcl-x(L) antagonism is sufficient to induce neuronal apoptosis, Bim likely promotes neuronal death by interacting with additional proteins besides Bcl-2/Bcl-x(L).


Subject(s)
Apoptosis/physiology , Carrier Proteins/biosynthesis , Membrane Proteins/biosynthesis , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Benzopyrans/pharmacology , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Count , Cells, Cultured , Dose-Response Relationship, Drug , Membrane Proteins/genetics , Membrane Proteins/physiology , Neurons/drug effects , Nitriles/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Sprague-Dawley , bcl-X Protein
14.
J Neurosci ; 24(44): 9993-10002, 2004 Nov 03.
Article in English | MEDLINE | ID: mdl-15525785

ABSTRACT

Glycogen synthase kinase-3beta (GSK-3beta) is a critical activator of neuronal apoptosis induced by a diverse array of neurotoxic insults. However, the downstream substrates of GSK-3beta that ultimately induce neuronal death are unknown. Here, we show that GSK-3beta phosphorylates and regulates the activity of Bax, a pro-apoptotic Bcl-2 family member that stimulates the intrinsic (mitochondrial) death pathway by eliciting cytochrome c release from mitochondria. In cerebellar granule neurons undergoing apoptosis, inhibition of GSK-3beta suppressed both the mitochondrial translocation of an expressed green fluorescent protein (GFP)-Bax(alpha) fusion protein and the conformational activation of endogenous Bax. GSK-3beta directly phosphorylated Bax(alpha) on Ser163, a residue found within a species-conserved, putative GSK-3beta phosphorylation motif. Coexpression of GFP-Bax(alpha) with a constitutively active mutant of GSK-3beta, GSK-3beta(Ser9Ala), enhanced the in vivo phosphorylation of wild-type Bax(alpha), but not a Ser163Ala mutant of Bax(alpha), in transfected human embryonic kidney 293 (HEK293) cells. Moreover, cotransfection with constitutively active GSK-3beta promoted the localization of Bax(alpha) to mitochondria and induced apoptosis in both transfected HEK293 cells and cerebellar granule neurons. In contrast, neither a Ser163Ala point mutant of Bax(alpha) nor a naturally occurring splice variant that lacks 13 amino acids encompassing Ser163 (Bax(sigma)) were driven to mitochondria in HEK293 cells coexpressing constitutively active GSK-3beta. In a similar manner, either mutation or deletion of the identified GSK-3beta phosphorylation motif prevented the localization of Bax to mitochondria in cerebellar granule neurons undergoing apoptosis. Our results indicate that GSK-3beta exerts some of its pro-apoptotic effects in neurons by regulating the mitochondrial localization of Bax, a key component of the intrinsic apoptotic cascade.


Subject(s)
Apoptosis/physiology , Glycogen Synthase Kinase 3/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cerebellum/cytology , Conserved Sequence , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/metabolism , Humans , Molecular Sequence Data , Neurons/metabolism , Neurons/ultrastructure , Phosphorylation , Protein Conformation , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Serine , bcl-2-Associated X Protein
15.
J Neurosci ; 24(19): 4498-509, 2004 May 12.
Article in English | MEDLINE | ID: mdl-15140920

ABSTRACT

The cellular mechanisms underlying Purkinje neuron death in various neurodegenerative disorders of the cerebellum are poorly understood. Here we investigate an in vitro model of cerebellar neuronal death. We report that cerebellar Purkinje neurons, deprived of trophic factors, die by a form of programmed cell death distinct from the apoptotic death of neighboring granule neurons. Purkinje neuron death was characterized by excessive autophagic-lysosomal vacuolation. Autophagy and death of Purkinje neurons were inhibited by nerve growth factor (NGF) and were activated by NGF-neutralizing antibodies. Although treatment with antisense oligonucleotides to the p75 neurotrophin receptor (p75ntr) decreased basal survival of cultured cerebellar neurons, p75ntr-antisense decreased autophagy and completely inhibited death of Purkinje neurons induced by trophic factor withdrawal. Moreover, adenoviral expression of a p75ntr mutant lacking the ligand-binding domain induced vacuolation and death of Purkinje neurons. These results suggest that p75ntr is required for Purkinje neuron survival in the presence of trophic support; however, during trophic factor withdrawal, p75ntr contributes to Purkinje neuron autophagy and death. The autophagic morphology resembles that found in neurodegenerative disorders, suggesting a potential role for this pathway in neurological disease.


Subject(s)
Adenine/analogs & derivatives , Autophagy/physiology , Cerebellum/cytology , Purkinje Cells/metabolism , Receptors, Nerve Growth Factor/physiology , Adenine/pharmacology , Animals , Animals, Newborn , Antibodies/pharmacology , Autophagy/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cells, Cultured , Lysosomes/metabolism , Lysosomes/pathology , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/pharmacology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuroprotective Agents/pharmacology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Purkinje Cells/drug effects , Purkinje Cells/pathology , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Vacuoles/drug effects , Vacuoles/pathology
16.
J Virol ; 78(12): 6360-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15163729

ABSTRACT

A common consequence of viral infection is perturbation of host cell nuclear functions. For cytoplasmically replicating viruses, this process may require regulated transport of specific viral proteins into the nucleus. Here, we describe a novel form of virus-induced perturbation of host cell nuclear structures. Active signal-mediated nuclear import of the reovirus sigma1s protein results in redistribution of nuclear pore complexes and nuclear lamins and formation of nuclear herniations. These herniations represent a previously undescribed mechanism by which cytoplasmic viral infection can perturb nuclear architecture and induce cytopathic effects, which ultimately lead to disease pathogenesis in the infected host.


Subject(s)
Active Transport, Cell Nucleus , Capsid Proteins/metabolism , Cell Nucleus/metabolism , Cell Nucleus/pathology , Mammalian orthoreovirus 3/pathogenicity , Capsid Proteins/genetics , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Lamina/pathology , Nuclear Localization Signals , Nuclear Pore/pathology , Sequence Analysis, DNA , Transfection
17.
J Neurochem ; 87(4): 914-21, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14622122

ABSTRACT

In vivo, the pesticide rotenone induces degeneration of dopamine neurons and parkinsonian-like pathology in adult rats. In the current study, we utilized primary ventral mesencephalic (VM) cultures from E15 rats as an in vitro model to examine the mechanism underlying rotenone-induced death of dopamine neurons. After 11 h of exposure to 30 nm rotenone, the number of dopamine neurons identified by tyrosine hydroxylase (TH) immunostaining declined rapidly with only 23% of the neurons surviving. By contrast, 73% of total cells survived rotenone treatment, indicating that TH+ neurons are more sensitive to rotenone. Examination of the role of apoptosis in TH+ neuron death, revealed that 10 and 30 nm rotenone significantly increased the number of apoptotic TH+ neurons from 7% under control conditions to 38 and 55%, respectively. The increase in apoptotic TH+ neurons correlated with an increase in immunoreactivity for active caspase-3 in TH+ neurons. The caspase-3 inhibitor, DEVD, rescued a significant number of TH+ neurons from rotenone-induced death. Furthermore, this protective effect lasted for at least 32 h post-rotenone and DEVD exposure, indicating lasting neuroprotection achieved with an intervention prior to the death commitment point. Our results show for the first time in primary dopamine neurons that, at low nanomolar concentrations, rotenone induces caspase-3-mediated apoptosis. Understanding the mechanism of rotenone-induced apoptosis in dopamine neurons may contribute to the development of new neuroprotective strategies against Parkinson's disease.


Subject(s)
Apoptosis , Caspases/metabolism , Mesencephalon , Neurons/drug effects , Pesticides/toxicity , Rotenone/toxicity , Animals , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Cells, Cultured , Dopamine/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Mesencephalon/cytology , Mesencephalon/embryology , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/biosynthesis
18.
J Biol Chem ; 278(42): 41472-81, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-12896970

ABSTRACT

Cerebellar granule neuron (CGN) survival depends on activity of the myocyte enhancer factor-2 (MEF2) transcription factors. Neuronal MEF2 activity is regulated by depolarization via a mechanism that is presently unclear. Here, we show that depolarization-mediated MEF2 activity and CGN survival are compromised by overexpression of the MEF2 repressor histone deacetylase-5 (HDAC5). Furthermore, removal of depolarization induced rapid cytoplasm-to-nuclear translocation of endogenous HDAC5. This effect was mimicked by addition of the calcium/calmodulin-dependent kinase (CaMK) inhibitor KN93 to depolarizing medium. Removal of depolarization or KN93 addition resulted in dephosphorylation of HDAC5 and its co-precipitation with MEF2D. HDAC5 nuclear translocation triggered by KN93 induced a marked loss of MEF2 activity and subsequent apoptosis. To selectively decrease CaMKII, CGNs were incubated with an antisense oligonucleotide to CaMKIIalpha. This antisense decreased CaMKIIalpha expression and induced nuclear shuttling of HDAC5 in CGNs maintained in depolarizing medium. Selectivity of the CaMKIIalpha antisense was demonstrated by its lack of effect on CaMKIV-mediated CREB phosphorylation. Finally, antisense to CaMKIIalpha induced caspase-3 activation and apoptosis, whereas a missense control oligonucleotide had no effect on CGN survival. These results indicate that depolarization-mediated calcium influx acts through CaMKII to inhibit HDAC5, thereby sustaining high MEF2 activity in CGNs maintained under depolarizing conditions.


Subject(s)
Cerebellum/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Histone Deacetylase Inhibitors , Neurons/metabolism , Transcription Factors/antagonists & inhibitors , Adenoviridae/genetics , Animals , Apoptosis , Blotting, Western , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspase 3 , Caspases/metabolism , Cell Nucleus/metabolism , Culture Media, Serum-Free/pharmacology , Cytoplasm/metabolism , Enzyme Activation , Epitopes , Genes, Dominant , Histone Deacetylases , Immunohistochemistry , MEF2 Transcription Factors , Mutation , Myogenic Regulatory Factors , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Potassium/pharmacology , Precipitin Tests , Protein Isoforms , Rats , Rats, Sprague-Dawley , Time Factors , Transcription, Genetic
19.
J Neurochem ; 85(6): 1488-99, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787068

ABSTRACT

Depolarization promotes the survival of cerebellar granule neurons via activation of the transcription factor myocyte enhancer factor 2D (MEF2D). Removal of depolarization induces hyperphosphorylation of MEF2D on serine/threonine residues, resulting in its decreased DNA binding and susceptibility to caspases. The subsequent loss of MEF2-dependent gene transcription contributes to the apoptosis of granule neurons. The kinase(s) that phosphorylates MEF2D during apoptosis is currently unknown. The serine/threonine kinase, glycogen synthase kinase-3 beta (GSK-3 beta), plays a pro-apoptotic role in granule neurons. To investigate a potential role for GSK-3 beta in MEF2D phosphorylation, we examined the effects of lithium, a non-competitive inhibitor of GSK-3 beta, on MEF2D activity in cultured cerebellar granule neurons. Lithium inhibited caspase-3 activation and chromatin condensation in granule neurons induced to undergo apoptosis by removal of depolarizing potassium and serum. Concurrently, lithium suppressed the hyperphosphorylation and caspase-mediated degradation of MEF2D. Moreover, lithium sustained MEF2 DNA binding and transcriptional activity in the absence of depolarization. Lithium also attenuated MEF2D hyperphosphorylation and apoptosis induced by calcineurin inhibition under depolarizing conditions, a GSK-3 beta-independent model of neuronal death. In contrast to lithium, MEF2D hyperphosphorylation was not inhibited by forskolin, insulin-like growth factor-I, or valproate, three mechanistically distinct inhibitors of GSK-3 beta. These results demonstrate that the kinase that phosphorylates and inhibits the pro-survival function of MEF2D in cerebellar granule neurons is a novel lithium target distinct from GSK-3 beta.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/metabolism , Lithium/pharmacology , Neurons/metabolism , Phosphotransferases/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Blood Proteins/pharmacology , Calcineurin Inhibitors , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cells, Cultured , Colforsin/pharmacology , DNA/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , MEF2 Transcription Factors , Myogenic Regulatory Factors , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Phosphotransferases/metabolism , Potassium/pharmacology , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
20.
J Neurosci ; 22(21): 9287-97, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12417654

ABSTRACT

Cerebellar granule neurons depend on insulin-like growth factor-I (IGF-I) for their survival. However, the mechanism underlying the neuroprotective effects of IGF-I is presently unclear. Here we show that IGF-I protects granule neurons by suppressing key elements of the intrinsic (mitochondrial) death pathway. IGF-I blocked activation of the executioner caspase-3 and the intrinsic initiator caspase-9 in primary cerebellar granule neurons deprived of serum and depolarizing potassium. IGF-I inhibited cytochrome c release from mitochondria and prevented its redistribution to neuronal processes. The effects of IGF-I on cytochrome c release were not mediated by blockade of the mitochondrial permeability transition pore, because IGF-I failed to inhibit mitochondrial swelling or depolarization. In contrast, IGF-I blocked induction of the BH3-only Bcl-2 family member, Bim (Bcl-2 interacting mediator of cell death), a mediator of Bax-dependent cytochrome c release. The suppression of Bim expression by IGF-I did not involve inhibition of the c-Jun transcription factor. Instead, IGF-I prevented activation of the forkhead family member, FKHRL1, another transcriptional regulator of Bim. Finally, adenoviral-mediated expression of dominant-negative AKT activated FKHRL1 and induced expression of Bim. These data suggest that IGF-I signaling via AKT promotes survival of cerebellar granule neurons by blocking the FKHRL1-dependent transcription of Bim, a principal effector of the intrinsic death-signaling cascade.


Subject(s)
Apoptosis/physiology , Carrier Proteins/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Membrane Proteins , Neurons/metabolism , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3 , Caspase 9 , Caspases/metabolism , Cells, Cultured , Cerebellum/cytology , Cytochrome c Group/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genes, Dominant , Humans , Insulin-Like Growth Factor I/physiology , JNK Mitogen-Activated Protein Kinases , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL