Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 28(5): 1627-1643, 2021 05.
Article in English | MEDLINE | ID: mdl-33277577

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra resulting in severe and progressive motor impairments. However, the mechanisms underlying this neuronal loss remain largely unknown. Oxidative stress and ER stress have been implicated in PD and these factors are known to activate the integrated stress response (ISR). Activating transcription factor 4 (ATF4), a key mediator of the ISR, and has been reported to induce the expression of genes involved in cellular homeostasis. However, during prolonged activation ATF4 can also induce the expression of pro-death target genes. Therefore, in the present study, we investigated the role of ATF4 in neuronal cell death in models of PD. We demonstrate that PD neurotoxins (MPP+ and 6-OHDA) and α-synuclein aggregation induced by pre-formed human alpha-synuclein fibrils (PFFs) cause sustained upregulation of ATF4 expression in mouse cortical and mesencephalic dopaminergic neurons. Furthermore, we demonstrate that PD neurotoxins induce the expression of the pro-apoptotic factors Chop, Trb3, and Puma in dopaminergic neurons in an ATF4-dependent manner. Importantly, we have determined that PD neurotoxin and α-synuclein PFF induced neuronal death is attenuated in ATF4-deficient dopaminergic neurons. Furthermore, ectopic expression of ATF4 but not transcriptionally defective ATF4ΔRK restores sensitivity of ATF4-deficient neurons to PD neurotoxins. Finally, we demonstrate that the eIF2α kinase inhibitor C16 suppresses MPP+ and 6-OHDA induced ATF4 activation and protects against PD neurotoxin induced dopaminergic neuronal death. Taken together these results indicate that ATF4 promotes dopaminergic cell death induced by PD neurotoxins and pathogenic α-synuclein aggregates and highlight the ISR factor ATF4 as a potential therapeutic target in PD.


Subject(s)
Activating Transcription Factor 4/metabolism , Neurotoxins/metabolism , Parkinson Disease/genetics , alpha-Synuclein/metabolism , Animals , Female , Humans , Male , Mice , Parkinson Disease/mortality , Survival Analysis
2.
Cell Immunol ; 332: 7-23, 2018 10.
Article in English | MEDLINE | ID: mdl-30017085

ABSTRACT

In response to micro-environmental cues such as microbial infections or T-helper 1 and 2 (TH1 and TH2) cytokines, macrophages (Mϕs) develop into M1- or M2-like phenotypes. Phenotypic polarization/activation of Mϕs are also essentially regulated by autocrine signals. Type-A γ-aminobutyric acid receptor (GABAAR)-mediated autocrine signaling is critical for phenotypic differentiation and transformation of various cell types. The present study explored whether GABAAR signaling regulates lung Mϕ (LMϕ) phenotypic activation under M1/TH1 and M2/TH2 environments. Results showed that GABAAR subunits were expressed by primary LMϕ of mice and the mouse Mϕ cell line RAW264.7. The expression levels of GABAAR subunits in mouse LMϕs and RAW264.7 cells decreased or increased concurrently with classical (M1) or alternative (M2) activation, respectively. Moreover, activation or blockade of GABAARs distinctively influenced the phenotypic characteristics of Mϕ. These results suggested that microenvironments leading to LMϕ phenotypic polarization concurrently modulates autocrine GABA signaling and its role in Mϕ activation.


Subject(s)
Autocrine Communication/physiology , Macrophage Activation/physiology , Macrophages, Alveolar/metabolism , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Cell Line , Cytokines/metabolism , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , RAW 264.7 Cells , Receptors, GABA/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism
4.
Mol Biol Cell ; 28(18): 2347-2359, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28701342

ABSTRACT

Pancreatitis is a debilitating disease of the exocrine pancreas that, under chronic conditions, is a major susceptibility factor for pancreatic ductal adenocarcinoma (PDAC). Although down-regulation of genes that promote the mature acinar cell fate is required to reduce injury associated with pancreatitis, the factors that promote this repression are unknown. Activating transcription factor 3 (ATF3) is a key mediator of the unfolded protein response, a pathway rapidly activated during pancreatic insult. Using chromatin immunoprecipitation followed by next-generation sequencing, we show that ATF3 is bound to the transcriptional regulatory regions of >30% of differentially expressed genes during the initiation of pancreatitis. Of importance, ATF3-dependent regulation of these genes was observed only upon induction of pancreatitis, with pathways involved in inflammation, acinar cell differentiation, and cell junctions being specifically targeted. Characterizing expression of transcription factors that affect acinar cell differentiation suggested that acinar cells lacking ATF3 maintain a mature cell phenotype during pancreatitis, a finding supported by maintenance of junctional proteins and polarity markers. As a result, Atf3-/- pancreatic tissue displayed increased tissue damage and inflammatory cell infiltration at early time points during injury but, at later time points, showed reduced acinar-to-duct cell metaplasia. Thus our results reveal a critical role for ATF3 as a key regulator of the acinar cell transcriptional response during injury and may provide a link between chronic pancreatitis and PDAC.


Subject(s)
Acinar Cells/metabolism , Activating Transcription Factor 3/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Acinar Cells/cytology , Activating Transcription Factor 3/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation/physiology , Ceruletide , Down-Regulation , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Phenotype , Pancreatic Neoplasms
5.
J Biol Chem ; 291(34): 17602-15, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27358397

ABSTRACT

Activation of Group I metabotropic glutamate receptors (mGluRs) activates signaling cascades, resulting in calcium release from intracellular stores, ERK1/2 activation, and long term changes in synaptic activity that are implicated in learning, memory, and neurodegenerative diseases. As such, elucidating the molecular mechanisms underlying Group I mGluR signaling is important for understanding physiological responses initiated by the activation of these receptors. In the current study, we identify the multifunctional scaffolding protein spinophilin as a novel Group I mGluR-interacting protein. We demonstrate that spinophilin interacts with the C-terminal tail and second intracellular loop of Group I mGluRs. Furthermore, we show that interaction of spinophilin with Group I mGluRs attenuates receptor endocytosis and phosphorylation of ERK1/2, an effect that is dependent upon the interaction of spinophilin with the C-terminal PDZ binding motif encoded by Group I mGluRs. Spinophilin knock-out results in enhanced mGluR5 endocytosis as well as increased ERK1/2, AKT, and Ca(2+) signaling in primary cortical neurons. In addition, the loss of spinophilin expression results in impaired mGluR5-stimulated LTD. Our results indicate that spinophilin plays an important role in regulating the activity of Group I mGluRs as well as their influence on synaptic activity.


Subject(s)
Calcium Signaling/physiology , Endocytosis/physiology , MAP Kinase Signaling System/physiology , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/metabolism , Animals , HEK293 Cells , Humans , Mice, Knockout , Microfilament Proteins/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Metabotropic Glutamate/genetics
6.
Mol Brain ; 8: 41, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26170135

ABSTRACT

Alzheimer's disease (AD) is characterized by the deposition of Beta-Amyloid (Aß) peptides in the brain. Aß peptides are generated by cleavage of the Amyloid Precursor Protein (APP) by the ß - and γ - secretase enzymes. Although this process is tightly linked to the internalization of cell surface APP, the compartments responsible are not well defined. We have found that APP can be rapidly internalized from the cell surface to lysosomes, bypassing early and late endosomes. Here we show by confocal microscopy and electron microscopy that this pathway is mediated by macropinocytosis. APP internalization is enhanced by antibody binding/crosslinking of APP suggesting that APP may function as a receptor. Furthermore, a dominant negative mutant of Arf6 blocks direct transport of APP to lysosomes, but does not affect classical endocytosis to endosomes. Arf6 expression increases through the hippocampus with the development of Alzheimer's disease, being expressed mostly in the CA1 and CA2 regions in normal individuals but spreading through the CA3 and CA4 regions in individuals with pathologically diagnosed AD. Disruption of lysosomal transport of APP reduces both Aß40 and Aß42 production by more than 30 %. Our findings suggest that the lysosome is an important site for Aß production and that altering APP trafficking represents a viable strategy to reduce Aß production.


Subject(s)
ADP-Ribosylation Factors/metabolism , Amyloid beta-Peptides/biosynthesis , Lysosomes/metabolism , Pinocytosis , ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factor 6 , Actins/metabolism , Alcohol Oxidoreductases/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cell Compartmentation , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Survival , Cross-Linking Reagents/metabolism , DNA-Binding Proteins/metabolism , Dextrans/metabolism , Endosomes/metabolism , Endosomes/ultrastructure , Gene Knockdown Techniques , Hippocampus/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/ultrastructure , Mice , Mutant Proteins/metabolism , Protein Transport , RNA, Small Interfering/metabolism , rac1 GTP-Binding Protein/metabolism
7.
Nat Commun ; 6: 6761, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25854456

ABSTRACT

How the brain's antioxidant defenses adapt to changing demand is incompletely understood. Here we show that synaptic activity is coupled, via the NMDA receptor (NMDAR), to control of the glutathione antioxidant system. This tunes antioxidant capacity to reflect the elevated needs of an active neuron, guards against future increased demand and maintains redox balance in the brain. This control is mediated via a programme of gene expression changes that boosts the synthesis, recycling and utilization of glutathione, facilitating ROS detoxification and preventing Puma-dependent neuronal apoptosis. Of particular importance to the developing brain is the direct NMDAR-dependent transcriptional control of glutathione biosynthesis, disruption of which can lead to degeneration. Notably, these activity-dependent cell-autonomous mechanisms were found to cooperate with non-cell-autonomous Nrf2-driven support from astrocytes to maintain neuronal GSH levels in the face of oxidative insults. Thus, developmental NMDAR hypofunction and glutathione system deficits, separately implicated in several neurodevelopmental disorders, are mechanistically linked.


Subject(s)
Electrical Synapses/metabolism , Frontal Lobe/metabolism , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Glutathione/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Dizocilpine Maleate/pharmacology , Electrical Synapses/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Frontal Lobe/drug effects , Gene Expression Regulation , Glutathione/drug effects , Glutathione Peroxidase/drug effects , Glutathione Transferase/drug effects , Mice , Mice, Knockout , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Transcription, Genetic/drug effects , Tumor Suppressor Proteins/genetics
8.
Expert Opin Ther Targets ; 18(11): 1293-304, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25118797

ABSTRACT

INTRODUCTION: Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion in the amino-terminal region of the huntingtin (htt) protein, which underlies the loss of striatal and cortical neurons. Glutamate has been implicated in a number of neurodegenerative diseases, and several studies suggest that the metabotropic glutamate receptor 5 (mGluR5) may represent a target for the treatment of HD. AREAS COVERED: The main goal of this review is to discuss the current data in the literature regarding the role of mGluR5 in HD and evaluate the potential of mGluR5 as a therapeutic target for the treatment of HD. mGluR5 is highly expressed in the brain regions affected in HD and is involved in movement control. Moreover, mGluR5 interacts with htt and mutated htt profoundly affects mGluR5 signaling. However, mGluR5 stimulation can activate both neuroprotective and neurotoxic signaling pathways, depending on the context of activation. EXPERT OPINION: Although the data published so far strongly indicate that mGluR5 plays a major role in HD-associated neurodegeneration, htt aggregation and motor symptoms, it is not clear whether mGluR5 stimulation can diminish or intensify neuronal cell loss and HD progression. Thus, future experiments will be necessary to further investigate the outcome of drugs acting on mGluR5 for the treatment of neurodegenerative diseases.


Subject(s)
Huntington Disease/drug therapy , Molecular Targeted Therapy , Receptor, Metabotropic Glutamate 5/drug effects , Animals , Brain/physiopathology , Drug Design , Glutamic Acid/metabolism , Humans , Huntingtin Protein , Huntington Disease/physiopathology , Nerve Tissue Proteins/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/drug effects
9.
Mol Brain ; 7: 40, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24886239

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) pathology occurs in part as the result of excessive production of ß-amyloid (Aß). Metabotropic glutamate receptor 5 (mGluR5) is now considered a receptor for Aß and consequently contributes to pathogenic Aß signaling in AD. RESULTS: Genetic deletion of mGluR5 rescues the spatial learning deficits observed in APPswe/PS1ΔE9 AD mice. Moreover, both Aß oligomer formation and Aß plaque number are reduced in APPswe/PS1ΔE9 mice lacking mGluR5 expression. In addition to the observed increase in Aß oligomers and plaques in APPswe/PS1ΔE9 mice, we found that both mTOR phosphorylation and fragile X mental retardation protein (FMRP) expression were increased in these mice. Genetic deletion of mGluR5 reduced Aß oligomers, plaques, mTOR phosphorylation and FMRP expression in APPswe/PS1ΔE9 mice. CONCLUSIONS: Thus, we propose that Aß activation of mGluR5 appears to initiate a positive feedback loop resulting in increased Aß formation and AD pathology in APPswe/PS1ΔE9 mice via mechanism that is regulated by FMRP.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Receptor, Metabotropic Glutamate 5/deficiency , Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Cell Membrane/metabolism , Cognition Disorders/complications , Cognition Disorders/physiopathology , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Gene Deletion , Humans , Inositol Phosphates/metabolism , Maze Learning , Memory Disorders/complications , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice , Mice, Knockout , Motor Activity , Phenotype , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
10.
Nat Commun ; 5: 3550, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24686499

ABSTRACT

Sustained cellular function and viability of high-energy demanding post-mitotic cells rely on the continuous supply of ATP. The utilization of mitochondrial oxidative phosphorylation for efficient ATP generation is a function of oxygen levels. As such, oxygen deprivation, in physiological or pathological settings, has profound effects on cell metabolism and survival. Here we show that mild extracellular acidosis, a physiological consequence of anaerobic metabolism, can reprogramme the mitochondrial metabolic pathway to preserve efficient ATP production regardless of oxygen levels. Acidosis initiates a rapid and reversible homeostatic programme that restructures mitochondria, by regulating mitochondrial dynamics and cristae architecture, to reconfigure mitochondrial efficiency, maintain mitochondrial function and cell survival. Preventing mitochondrial remodelling results in mitochondrial dysfunction, fragmentation and cell death. Our findings challenge the notion that oxygen availability is a key limiting factor in oxidative metabolism and brings forth the concept that mitochondrial morphology can dictate the bioenergetic status of post-mitotic cells.


Subject(s)
Acidosis/metabolism , Acidosis/physiopathology , Mitochondria/metabolism , Oxygen/metabolism , Acidosis/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Cell Survival , Female , Humans , Male , Metabolic Networks and Pathways , Mice , Mitosis , Oxidative Phosphorylation
11.
Hum Mol Genet ; 23(8): 2030-42, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24282028

ABSTRACT

Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder caused by a polyglutamine expansion in the amino-terminal region of the huntingtin protein, which promotes progressive neuronal cell loss, neurological symptoms and death. In the present study, we show that blockade of mGluR5 with MTEP promotes increased locomotor activity in both control (Hdh(Q20/Q20)) and mutant HD (Hdh(Q111/Q111)) mice. Although acute injection of MTEP increases locomotor activity in both control and mutant HD mice, locomotor activity is increased in only control mice, not mutant HD mice, following the genetic deletion of mGluR5. Interestingly, treatment of mGluR5 knockout mice with either D1 or D2 dopamine antagonists eliminates the increased locomotor activity of mGluR5 knockout mice. Amphetamine treatment increases locomotor activity in control mice, but not mGluR5 null mutant HD mice. However, the loss of mGluR5 expression improves rotarod performance and decreases the number of huntingtin intranuclear inclusions in mutant HD mice. These adaptations may be due to mutant huntingtin-dependent alterations in gene expression, as microarray studies have identified several genes that are altered in mutant, but not wild-type HD mice lacking mGluR5 expression. qPCR experiments confirm that the mRNA transcript levels of dynein heavy chain, dynactin 3 and dynein light chain-6 are altered following the genetic deletion of mGluR5 in mutant HD mice, as compared with wild-type mutant HD mice. Thus, our data suggest that mutant huntingtin protein and mGluR5 exhibit a functional interaction that may be important for HD-mediated alterations in locomotor behavior and the development of intranuclear inclusions.


Subject(s)
Disease Models, Animal , Huntington Disease/pathology , Intranuclear Inclusion Bodies/pathology , Motor Activity/physiology , Receptor, Metabotropic Glutamate 5/physiology , Serotonin Plasma Membrane Transport Proteins/physiology , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Gene Expression Profiling , Huntington Disease/genetics , Huntington Disease/metabolism , Immunoenzyme Techniques , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/metabolism , Mice , Mice, Knockout , Motor Activity/drug effects , Pyridines/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/pharmacology
12.
Mol Cell ; 50(4): 565-76, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706821

ABSTRACT

Although Numb exhibits its tumor-suppressive function in breast cancer in part by binding to and stabilizing p53, it is unknown how the Numb-p53 interaction is regulated in cells. We found that Numb is methylated in its phosphotyrosine-binding (PTB) domain by the lysine methyltransferase Set8. Moreover, methylation uncouples Numb from p53, resulting in increased p53 ubiquitination and degradation. While Numb promotes apoptosis in a p53-dependent manner, the apoptotic function is abolished when Numb is methylated by Set8 or the Lys methylation sites in Numb are mutated. Conversely, the Numb-p53 interaction and Numb-mediated apoptosis are significantly enhanced by depletion of Set8 from cancer cells or by treating the cells with doxorubicin, a chemotherapeutic drug that causes a reduction in the mRNA and protein levels of Set8. Our work identifies the Set8-Numb-p53 signaling axis as an important regulatory pathway for apoptosis and suggests a therapeutic strategy by targeting Numb methylation.


Subject(s)
Apoptosis/physiology , Histone-Lysine N-Methyltransferase/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Binding Sites/genetics , Cell Line , Cell Line, Tumor , Doxorubicin/pharmacology , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Immunoblotting , Lysine/genetics , Lysine/metabolism , MCF-7 Cells , Membrane Proteins/genetics , Methylation , Microscopy, Confocal , Mutation , Nerve Tissue Proteins/genetics , Protein Binding , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics
13.
Mol Brain ; 6: 9, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23406666

ABSTRACT

BACKGROUND: Group I metabotropic glutamate receptors (mGluR) are coupled via Gαq/11 to the activation of phospholipase Cß, which hydrolyzes membrane phospholipids to form inositol 1,4,5 trisphosphate and diacylglycerol. In addition to functioning as neurotransmitter receptors to modulate synaptic activity, pathological mGluR5 signaling has been implicated in a number of disease processes including Fragile X, amyotrophic lateral sclerosis, multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, and drug addiction. The expression of mGluR5 in astrocytes has been shown to be increased in several acute and chronic neurodegenerative conditions, but little is known about the functional relevance of mGluR5 up-regulation in astrocytes following injury. RESULTS: In the current study, we investigated primary mouse cortical astrocyte cell death in response to oxygen glucose deprivation (OGD) and found that OGD induced both necrotic and apoptotic cell death of astrocytes. OGD resulted in an increase in astrocytic mGluR5 protein expression, inositol phosphate formation and extracellular regulated kinase (ERK1/2) phosphorylation, but only inositol phosphate formation was blocked with the mGluR5 selective antagonist MPEP. Cortical astrocytes derived from mGluR5 knockout mice exhibited resistance to OGD-stimulated apoptosis, but a lack of mGluR5 expression did not confer protection against necrotic cell death. The antagonism of the inositol 1,4,5 trisphosphate receptor also reduced apoptotic cell death in wild-type astrocytes, but did not provide any additional protection to astrocytes derived from mGluR5 null mice. Moreover, the disruption of Homer protein interactions with mGluR5 also reduced astrocyte apoptosis. CONCLUSION: Taken together these observations indicated that mGluR5 up-regulation contributed selectively to the apoptosis of astrocytes via the activation of phospholipase C and the release of calcium from intracellular stores as well as via the association with Homer proteins.


Subject(s)
Apoptosis/drug effects , Astrocytes/metabolism , Carrier Proteins/metabolism , Glucose/deficiency , Oxygen/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Astrocytes/pathology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose/pharmacology , Homer Scaffolding Proteins , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol Phosphates/metabolism , Mice , Necrosis , Phosphorylation/drug effects , Receptor, Metabotropic Glutamate 5
14.
PLoS One ; 7(10): e46885, 2012.
Article in English | MEDLINE | ID: mdl-23056511

ABSTRACT

The AKT, GSK3 and JNK family kinases have been implicated in neuronal apoptosis associated with neuronal development and several neurodegenerative conditions. However, the mechanisms by which these kinase pathways regulate apoptosis remain unclear. In this study we have investigated the role of these kinases in neuronal cell death using an established model of trophic factor deprivation induced apoptosis in cerebellar granule neurons. BCL-2 family proteins are known to be central regulators of apoptosis and we have determined that the pro-apoptotic family member Puma is transcriptionally up-regulated in trophic factor deprived neurons and that Puma induction is required for apoptosis in vitro and in vivo. Importantly, we demonstrate that Puma induction is dependent on both JNK activation and AKT inactivation. AKT is known to regulate a number of downstream pathways, however we have determined that PI3K-AKT inactivation induces Puma expression through a GSK3ß-dependent mechanism. Finally we demonstrate that the JNK and AKT/GSK3ß pathways converge to regulate FoxO3a-mediated transcriptional activation of Puma. In summary we have identified a novel and critical link between the AKT, GSK3ß and JNK kinases and the regulation of Puma induction and suggest that this may be pivotal to the regulation of neuronal apoptosis in neurodegenerative conditions.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis , Glycogen Synthase Kinase 3/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Animals , Enzyme Activation , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Neurons/metabolism , Potassium/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Transcriptional Activation
15.
J Neurosci ; 30(50): 16938-48, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159964

ABSTRACT

An increasing body of evidence points to a key role of endoplasmic reticulum (ER) stress in acute and chronic neurodegenerative conditions. Extensive ER stress can trigger neuronal apoptosis, but the signaling pathways that regulate this cell death remain unclear. In the present study, we demonstrate that PUMA, a Bcl-2 homology 3 (BH3)-only member of the Bcl-2 family, is transcriptionally activated in cortical neurons by ER stress and is essential for ER-stress-induced cell death. PUMA is known to be a key transcriptional target of p53, but we have found that ER stress triggers PUMA induction and cell death through a p53-independent mechanism mediated by the ER-stress-inducible transcription factor ATF4 (activating transcription factor 4). Specifically, we demonstrate that ectopic expression of ATF4 sensitizes mouse cortical neurons to ER-stress-induced apoptosis and that ATF4-deficient neurons exhibit markedly reduced levels of PUMA expression and cell death. However, chromatin immunoprecipitation experiments suggest that ATF4 does not directly regulate the PUMA promoter. Rather, we found that ATF4 induces expression of the transcription factor CHOP (C/EBP homologous protein) and that CHOP in turn activates PUMA induction. Specifically, we demonstrate that CHOP binds to the PUMA promoter during ER stress and that CHOP knockdown attenuates PUMA induction and neuronal apoptosis. In summary, we have identified a key signaling pathway in ER-stress-induced neuronal death involving ATF4-CHOP-mediated transactivation of the proapoptotic Bcl-2 family member PUMA. We propose that this pathway may be an important therapeutic target relevant to a number of neurodegenerative conditions.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis/genetics , Endoplasmic Reticulum/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Stress, Physiological/genetics , Transcription Factor CHOP/metabolism , Tumor Suppressor Proteins/metabolism , Activating Transcription Factor 4/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Cell Culture Techniques , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Endoplasmic Reticulum/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Knockout , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Stress, Physiological/physiology , Thapsigargin/pharmacology , Transcription Factor CHOP/genetics , Transfection/methods , Tumor Suppressor Proteins/genetics , Tunicamycin/pharmacology
16.
CNS Neurol Disord Drug Targets ; 9(5): 574-95, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20632969

ABSTRACT

Stimulation of Group I metabotropic glutamate receptors (mGluR1 and mGluR5) leads to activation of a wide variety of signalling pathways. mGluRs couple to Gα(q/)11 proteins, activating phospholipase Cß1 resulting in both diacylglycerol and inositol-1,4,5-triphosphate formation followed by the activation of protein kinase C. In addition, mGluR activation can lead to modulation of a number of ion channels, such as different types of calcium and potassium channels. Group I mGluRs can also activate other downstream protein kinases, such as ERK1/2 and AKT, which are implicated in cellular growth, differentiation, and survival. Moreover, Group I mGluRs interact with a variety of different proteins that are important for the regulation of synaptic signalling, such as Homer and PDZ domain containing proteins, such as Tamalin. A role for mGluR1/5 in a number of disease states has also been proposed. As mGluR1/5 signal transduction is complex and involves multiple partners, a better understanding of alterations in mGluR signalling in brain disorders will be required in order to discern the molecular and cellular basis of these pathologies. This review will highlight recent findings concerning mGluR signaling alterations in brain pathologies, such as stroke, fragile X syndrome, Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, and drug addiction.


Subject(s)
Brain Diseases/metabolism , Nervous System Diseases/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Allosteric Regulation/drug effects , Animals , Humans , Models, Biological , Nervous System Diseases/physiopathology , Receptor, Metabotropic Glutamate 5 , Signal Transduction/physiology
17.
Mol Brain ; 3: 11, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20409323

ABSTRACT

BACKGROUND: A central feature of Alzheimer's disease is the cleavage of the amyloid precursor protein (APP) to form beta-amyloid peptide (Abeta) by the beta-secretase and gamma-secretase enzymes. Although this has been shown to occur after endocytosis of APP from the cell surface, the exact compartments of APP processing are not well defined. We have previously demonstrated that APP and gamma-secretase proteins and activity are highly enriched in purified rat liver lysosomes. In order to examine the lysosomal distribution and trafficking of APP in cultured cells, we generated constructs containing APP fused to a C-terminal fluorescent protein tag and N-terminal HA-epitope tag. These were co-transfected with a panel of fluorescent-protein tagged compartment markers. RESULTS: Here we demonstrate using laser-scanning confocal microscopy that although APP is present throughout the endosomal/lysosomal system in transfected Cos7 and neuronal SN56 cell lines as well as in immunostained cultured mouse neurons, it is enriched in the lysosome. We also show that the Swedish and London mutations reduce the amount of APP in the lysosome. Surprisingly, in addition to its expected trafficking from the cell surface to the early and then late endosomes, we find that cell-surface labelled APP is transported rapidly and directly from the cell surface to lysosomes in both Cos7 and SN56 cells. This rapid transit to the lysosome is blocked by the presence of either the London or Swedish mutations. CONCLUSIONS: These results demonstrate the presence of a novel, rapid and specific transport pathway from the cell surface to the lysosomes. This suggests that regulation of lysosomal traffic could regulate APP processing and that the lysosome could play a central role in the pathophysiology of Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Membrane/metabolism , Lysosomes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Biological Transport/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Mice , Mutation , Neurons/cytology , Neurons/metabolism , Rats
18.
J Neurosci ; 30(11): 3973-82, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20237268

ABSTRACT

Developmental and pathological death of neurons requires activation of a defined pathway of cell cycle proteins. However, it is unclear how this pathway is regulated and whether it is relevant in vivo. A screen for transcripts robustly induced in cultured neurons by DNA damage identified Sertad1, a Cdk4 (cyclin-dependent kinase 4) activator. Sertad1 is also induced in neurons by nerve growth factor (NGF) deprivation and Abeta (beta-amyloid). RNA interference-mediated downregulation of Sertad1 protects neurons in all three death models. Studies of NGF withdrawal indicate that Sertad1 is required to initiate the apoptotic cell cycle pathway since its knockdown blocks subsequent pathway events. Finally, we find that Sertad1 expression is required for developmental neuronal death in the cerebral cortex. Sertad1 thus appears to be essential for neuron death in trophic support deprivation in vitro and in vivo and in models of DNA damage and Alzheimer's disease. It may therefore be a suitable target for therapeutic intervention.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Neurons/pathology , Neurons/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Cell Survival/genetics , Cell Survival/physiology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Cyclin-Dependent Kinase 4/metabolism , DNA Damage/genetics , Enzyme Activation/genetics , Gene Expression Regulation, Developmental/physiology , Mice , Neurons/enzymology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , PC12 Cells , RNA Interference/physiology , Rats , Rats, Sprague-Dawley , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcription Factors
19.
J Neurosci ; 30(1): 316-24, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053912

ABSTRACT

Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder caused by a polyglutamine expansion in the huntingtin protein (Htt). Group I metabotropic glutamate receptors (mGluRs) are coupled to G(alphaq) and play an important role in neuronal survival. We have previously demonstrated that mGluRs interact with Htt. Here we used striatal neuronal primary cultures and acute striatal slices to demonstrate that mGluR-mediated signaling pathways are altered in a presymptomatic mouse model of HD (Hdh(Q111/Q111)), as compared to those of control mice (Hdh(Q20/Q20)). mGluR1/5-mediated inositol phosphate (InsP) formation is desensitized in striatal slices from Hdh(Q111/Q111) mice and this desensitization is PKC-mediated. Despite of decreased InsP formation, (S)-3,5-dihydroxylphenylglycine (DHPG)-mediated Ca(2+) release is higher in Hdh(Q111/Q111) than in Hdh(Q20/Q20) neurons. Furthermore, mGluR1/5-stimulated AKT and extracellular signal-regulated kinase (ERK) activation is altered in Hdh(Q111/Q111) mice. Basal AKT activation is higher in Hdh(Q111/Q111) neurons and this increase is mGluR5 dependent. Moreover, mGluR5 activation leads to higher levels of ERK activation in Hdh(Q111/Q111) than in Hdh(Q20/Q20) striatum. PKC inhibition not only brings Hdh(Q111/Q111) DHPG-stimulated InsP formation to Hdh(Q20/Q20) levels, but also causes an increase in neuronal cell death in Hdh(Q111/Q111) neurons. However, PKC inhibition does not modify neuronal cell death in Hdh(Q20/Q20) neurons, suggesting that PKC-mediated desensitization of mGluR1/5 in Hdh(Q111/Q111) mice might be protective in HD. Together, these data indicate that group I mGluR-mediated signaling pathways are altered in HD and that these cell signaling adaptations could be important for striatal neurons survival.


Subject(s)
Disease Models, Animal , Huntington Disease/physiopathology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Gene Knock-In Techniques , Mice , Mice, Transgenic
20.
J Neurochem ; 112(2): 497-510, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19895669

ABSTRACT

DNA damage is a critical component of neuronal death underlying neurodegenerative diseases and injury. Neuronal death evoked by DNA damage is characterized by inappropriate activation of multiple cell cycle components. However, the mechanism regulating this activation is not fully understood. We demonstrated previously that the cell division cycle (Cdc) 25A phosphatase mediates the activation of cyclin-dependent kinases and neuronal death evoked by the DNA damaging agent camptothecin. We also showed that Cdc25A activation is blocked by constitutive checkpoint kinase 1 activity under basal conditions in neurons. Presently, we report that an additional factor is central to regulation of Cdc25A phosphatase in neuronal death. In a gene array screen, we first identified Pim-1 as a potential factor up-regulated following DNA damage. We confirmed the up-regulation of Pim-1 transcript, protein and kinase activity following DNA damage. This induction of Pim-1 is regulated by the nuclear factor kappa beta (NF-kappaB) pathway as Pim-1 expression and activity are significantly blocked by siRNA-mediated knockdown of NF-kappaB or NF-kappaB pharmacological inhibitors. Importantly, Pim-1 activity is critical for neuronal death in this paradigm and its deficiency blocks camptothecin-mediated neuronal death. It does so by activating Cdc25A with consequent activation of cyclin D1-associated kinases. Taken together, our results demonstrate that Pim-1 kinase plays a central role in DNA damage-evoked neuronal death by regulating aberrant neuronal cell cycle activation.


Subject(s)
Cell Cycle/physiology , DNA Damage/physiology , Neurons/physiology , Proto-Oncogene Proteins c-pim-1/metabolism , Adenosine Triphosphate/metabolism , Animals , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Line, Transformed , Cerebral Cortex/cytology , Chromatin Immunoprecipitation/methods , DNA Damage/drug effects , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Knockout , NF-kappa B/metabolism , Neurons/drug effects , Proto-Oncogene Proteins c-pim-1/deficiency , Proto-Oncogene Proteins c-pim-1/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Staurosporine/pharmacology , Sulfur Isotopes/metabolism , Time Factors , Transfection/methods , Up-Regulation/drug effects , Up-Regulation/physiology , cdc25 Phosphatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...