Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 9(403)2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28814543

ABSTRACT

Hallmarks of chronic neurodegenerative disease include progressive synaptic loss and neuronal cell death, yet the cellular pathways that underlie these processes remain largely undefined. We provide evidence that dual leucine zipper kinase (DLK) is an essential regulator of the progressive neurodegeneration that occurs in amyotrophic lateral sclerosis and Alzheimer's disease. We demonstrate that DLK/c-Jun N-terminal kinase signaling was increased in mouse models and human patients with these disorders and that genetic deletion of DLK protected against axon degeneration, neuronal loss, and functional decline in vivo. Furthermore, pharmacological inhibition of DLK activity was sufficient to attenuate the neuronal stress response and to provide functional benefit even in the presence of ongoing disease. These findings demonstrate that pathological activation of DLK is a conserved mechanism that regulates neurodegeneration and suggest that DLK inhibition may be a potential approach to treat multiple neurodegenerative diseases.


Subject(s)
Leucine Zippers , MAP Kinase Kinase Kinases/metabolism , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Signal Transduction , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Gene Deletion , Gene Expression Regulation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Mice, Transgenic , Neuroprotection , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Spinal Cord/enzymology , Spinal Cord/pathology , Superoxide Dismutase/metabolism
2.
J Exp Med ; 210(12): 2553-67, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24166713

ABSTRACT

Excessive glutamate signaling is thought to underlie neurodegeneration in multiple contexts, yet the pro-degenerative signaling pathways downstream of glutamate receptor activation are not well defined. We show that dual leucine zipper kinase (DLK) is essential for excitotoxicity-induced degeneration of neurons in vivo. In mature neurons, DLK is present in the synapse and interacts with multiple known postsynaptic density proteins including the scaffolding protein PSD-95. To examine DLK function in the adult, DLK-inducible knockout mice were generated through Tamoxifen-induced activation of Cre-ERT in mice containing a floxed DLK allele, which circumvents the neonatal lethality associated with germline deletion. DLK-inducible knockouts displayed a modest increase in basal synaptic transmission but had an attenuation of the JNK/c-Jun stress response pathway activation and significantly reduced neuronal degeneration after kainic acid-induced seizures. Together, these data demonstrate that DLK is a critical upstream regulator of JNK-mediated neurodegeneration downstream of glutamate receptor hyper-activation and represents an attractive target for the treatment of indications where excitotoxicity is a primary driver of neuronal loss.


Subject(s)
MAP Kinase Kinase Kinases/physiology , Nerve Degeneration/physiopathology , Animals , Brain/pathology , Brain/physiopathology , Disks Large Homolog 4 Protein , Glutamic Acid/physiology , Guanylate Kinases/physiology , Kainic Acid/toxicity , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System , Membrane Proteins/physiology , Mice , Mice, Knockout , N-Methylaspartate/physiology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Tissue Proteins/physiology , Synapses/physiology
3.
J Cell Biol ; 202(5): 747-63, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-23979718

ABSTRACT

Neurons are highly polarized cells that often project axons a considerable distance. To respond to axonal damage, neurons must transmit a retrograde signal to the nucleus to enable a transcriptional stress response. Here we describe a mechanism by which this signal is propagated through injury-induced stabilization of dual leucine zipper-bearing kinase (DLK/MAP3K12). After neuronal insult, specific sites throughout the length of DLK underwent phosphorylation by c-Jun N-terminal kinases (JNKs), which have been shown to be downstream targets of DLK pathway activity. These phosphorylation events resulted in increased DLK abundance via reduction of DLK ubiquitination, which was mediated by the E3 ubiquitin ligase PHR1 and the de-ubiquitinating enzyme USP9X. Abundance of DLK in turn controlled the levels of downstream JNK signaling and apoptosis. Through this feedback mechanism, the ubiquitin-proteasome system is able to provide an additional layer of regulation of retrograde stress signaling to generate a global cellular response to localized external insults.


Subject(s)
Apoptosis , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Sensory Receptor Cells/enzymology , Sensory Receptor Cells/pathology , Ubiquitination , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/pathology , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Enzyme Stability/drug effects , HEK293 Cells , Humans , Mice , Models, Biological , Molecular Weight , Nerve Crush , Nerve Growth Factor/pharmacology , Optic Nerve/drug effects , Optic Nerve/pathology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Stress, Physiological/drug effects , Ubiquitin/metabolism , Ubiquitination/drug effects
4.
Nat Chem Biol ; 8(7): 655-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22683611

ABSTRACT

The caspases are a family of cytosolic proteases with essential roles in inflammation and apoptosis. Drug discovery efforts have focused on developing molecules directed against the active sites of caspases, but this approach has proved challenging and has not yielded any approved therapeutics. Here we describe a new strategy for generating inhibitors of caspase-6, a potential therapeutic target in neurodegenerative disorders, by screening against its zymogen form. Using phage display to discover molecules that bind the zymogen, we report the identification of a peptide that specifically impairs the function of caspase-6 in vitro and in neuronal cells. Remarkably, the peptide binds at a tetramerization interface that is uniquely present in zymogen caspase-6, rather than binding into the active site, and acts via a new allosteric mechanism that promotes caspase tetramerization. Our data illustrate that screening against the zymogen holds promise as an approach for targeting caspases in drug discovery.


Subject(s)
Biopolymers/metabolism , Caspase 6/metabolism , Enzyme Precursors/metabolism , Peptides/metabolism , Allosteric Regulation , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Peptides/chemistry , Protein Binding
5.
PLoS One ; 7(1): e30376, 2012.
Article in English | MEDLINE | ID: mdl-22253931

ABSTRACT

Caspase-6 is a cysteinyl protease implicated in neurodegenerative conditions including Alzheimer's and Huntington's disease making it an attractive target for therapeutic intervention. A greater understanding of the role of caspase-6 in disease has been hampered by a lack of suitable cellular assays capable of specifically detecting caspase-6 activity in an intact cell environment. This is mainly due to the use of commercially available peptide substrates and inhibitors which lack the required specificity to facilitate development of this type of assay. We report here a 384-well whole-cell chemiluminescent ELISA assay that monitors the proteolytic degradation of endogenously expressed lamin A/C during the early stages of caspase-dependent apoptosis. The specificity of lamin A/C proteolysis by caspase-6 was demonstrated against recombinant caspase family members and further confirmed in genetic deletion studies. In the assay, plasma membrane integrity remained intact as assessed by release of lactate dehydrogenase from the intracellular environment and the exclusion of cell impermeable peptide inhibitors, despite the induction of an apoptotic state. The method described here is a robust tool to support drug discovery efforts targeting caspase-6 and is the first reported to specifically monitor endogenous caspase-6 activity in a cellular context.


Subject(s)
Biological Assay/methods , Caspase 6/metabolism , Cells/enzymology , Enzyme Assays/methods , Lamin Type A/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Blotting, Western , Caspase Inhibitors , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells/drug effects , Enzyme Activation/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Mice , Molecular Sequence Data , Peptides/chemistry , Peptides/pharmacology , Protease Inhibitors/pharmacology , Protein Isoforms/metabolism , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/metabolism , Staurosporine/pharmacology , Substrate Specificity/drug effects
6.
J Cell Biol ; 194(5): 751-64, 2011 Sep 05.
Article in English | MEDLINE | ID: mdl-21893599

ABSTRACT

The c-Jun N-terminal kinase (JNK) signaling pathway is essential for neuronal degeneration in multiple contexts but also regulates neuronal homeostasis. It remains unclear how neurons are able to dissociate proapoptotic JNK signaling from physiological JNK activity. In this paper, we show that the mixed lineage kinase dual leucine zipper kinase (DLK) selectively regulates the JNK-based stress response pathway to mediate axon degeneration and neuronal apoptosis without influencing other aspects of JNK signaling. This specificity is dependent on interaction of DLK with the scaffolding protein JIP3 to form a specialized JNK signaling complex. Local activation of DLK-based signaling in the axon results in phosphorylation of c-Jun and apoptosis after redistribution of JNK to the cell body. In contrast, regulation of axon degeneration by DLK is c-Jun independent and mediated by distinct JNK substrates. DLK-null mice displayed reduced apoptosis in multiple neuronal populations during development, demonstrating that prodegenerative DLK signaling is required in vivo.


Subject(s)
Apoptosis/physiology , Central Nervous System/embryology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/drug effects , Axons/metabolism , Axons/pathology , Axons/physiology , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/metabolism , Central Nervous System/pathology , Embryo, Mammalian/embryology , Embryo, Mammalian/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/pathology , Homeodomain Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Nerve Growth Factor/deficiency , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Binding/physiology , Protein Kinase Inhibitors/pharmacokinetics , Protein Transport/physiology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/genetics , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord/pathology , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Proc Natl Acad Sci U S A ; 107(50): 21795-800, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098272

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are lineage-restricted progenitors generally limited in vivo to producing oligodendrocytes. Mechanisms controlling genesis of OPCs are of interest because of their importance in myelin development and their potential for regenerative therapies in multiple sclerosis and dysmyelinating syndromes. We show here that the SoxE transcription factors (comprising Sox8, 9, and 10) induce multipotent neural precursor cells (NPCs) from the early postnatal subventricular zone (SVZ) to become OPCs in an autonomous manner. We performed a chromatin immunoprecipitation-based bioinformatic screen and identified Suppressor of Fused (Sufu) as a direct target of repression by Sox10. In vitro, overexpression of Sufu blocked OPC production, whereas RNAi-mediated inhibition augmented OPC production. Furthermore, mice heterozygous for Sufu have increased numbers of OPCs in the telencephalon during development. We conclude that Sox10 acts to restrict the potential of NPCs toward the oligodendrocyte lineage in part by regulating the expression of Sufu.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , Neural Stem Cells/physiology , Neurogenesis/physiology , Oligodendroglia/physiology , Repressor Proteins/metabolism , SOXE Transcription Factors/metabolism , Animals , Cells, Cultured , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Neural Stem Cells/cytology , Oligodendroglia/cytology , Repressor Proteins/genetics , SOXE Transcription Factors/genetics
8.
J Neurosci ; 30(40): 13367-72, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926663

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are generated from multiple progenitor domains in the telencephalon in developmental succession from ventral to dorsal. Previous studies showed that Wnt signaling inhibits the differentiation of OPCs into mature oligodendrocytes. Here we explored the hypothesis that Wnt signaling limits the generation of OPCs from neural progenitors during forebrain development. We manipulated Wnt signaling in mouse neural progenitor cultures and found that Wnt signaling influences progenitors cell autonomously to alter the production of OPCs, and that endogenous Wnt signaling in these cultures limits the efficiency of generating OPCs from neural progenitors. To examine these events in vivo, we electroporated a soluble Wnt inhibitor or a dominant-negative transcriptional regulator into embryonic mouse neocortical ventricular zone before the usual onset of OPC production and showed that decreasing Wnt signaling in cortical progenitors results in early production of OPCs. Our studies indicate that Wnt signaling influences the timing and extent of OPC production in the developing telencephalon.


Subject(s)
Cell Differentiation/physiology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism , Telencephalon/embryology , Wnt Proteins/physiology , Animals , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Electroporation , Female , Lateral Ventricles/cytology , Lateral Ventricles/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/cytology , Neocortex/embryology , Signal Transduction/genetics , Telencephalon/cytology , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt3 Protein
9.
Genome Biol ; 7(3): 207, 2006.
Article in English | MEDLINE | ID: mdl-16584531

ABSTRACT

Adult neurogenesis in the hippocampus is under complex genetic control. A recent comparative study of two inbred mouse strains using quantitative trait locus analysis has revealed that cell survival is most highly correlated with neurogenesis and identified candidate genes for further investigation.


Subject(s)
Hippocampus/growth & development , Mice, Inbred BALB C/genetics , Mice, Inbred C57BL/genetics , Neurons/physiology , Quantitative Trait Loci , Animals , Humans , Mice
10.
Sci STKE ; 2006(319): pe5, 2006 Jan 24.
Article in English | MEDLINE | ID: mdl-16434726

ABSTRACT

It is now widely accepted that discrete regions of the adult brain contain stem cells that continue to generate new neurons. However, it remains unclear what molecular signals define the neurogenic niche and how such signals act on the heterogeneous cell populations within these regions. Here we discuss two recent studies that demonstrate the role of Wnt and Sonic Hedgehog signaling in neurogenic zones. Wnts act on neuronal precursors that mature and contribute to the dentate gyrus (DG), whereas Sonic Hedgehog affects the bona fide stem cells and transit amplifying cells (the partially committed progeny of stem cells). These studies further define how discrete populations of cells react to specific extracellular signals provided within the neurogenic niche to survive, proliferate, and form functional mature cell types.


Subject(s)
Dentate Gyrus/cytology , Neurons/cytology , Signal Transduction/physiology , Stem Cells/cytology , Trans-Activators/physiology , Wnt Proteins/physiology , Animals , Astrocytes/cytology , Cell Differentiation/physiology , Cell Lineage , Hedgehog Proteins , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Models, Neurological , Morphogenesis/physiology , Oligodendroglia/cytology , Rats , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/physiology , Tamoxifen/pharmacology , Zinc Finger Protein GLI1
11.
J Neurosci ; 22(22): 9800-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12427836

ABSTRACT

Here, we show that the p53 family member, p73, is necessary for survival and long-term maintenance of CNS neurons, including postnatal cortical neurons. In p73-/- animals, cortical neuron number is normal at birth but decreases significantly by postnatal day 14 (P14)-P16 because of enhanced apoptosis. This decrease continues into adulthood, when p73-/- animals have approximately one-half as many cortical cells as their wild-type littermates. Cortical neurons express the DeltaNp73alpha protein, and overexpression of DeltaNp73 isoforms rescues cortical neurons from diverse apoptotic stimuli. Thus, DeltaNp73 isoforms are survival proteins in cortical neurons, and their deletion causes a gradual loss of cortical neurons in the weeks and months after birth. This decrease in CNS neuron number in p73-/- animals is not limited to the cortex; facial motor neuron number is decreased, and postnatal development of the olfactory bulb is greatly perturbed. These findings, together with our previous work showing that DeltaNp73 is essential for survival of peripheral sympathetic neurons (Pozniak et al., 2000), indicate that p73 isoforms are essential survival proteins in CNS as well as PNS neurons, and that they likely play a role not only during developmental cell death but also in the long-term maintenance of at least some adult neurons.


Subject(s)
Central Nervous System/pathology , DNA-Binding Proteins/deficiency , Neurodegenerative Diseases/pathology , Neurons/pathology , Nuclear Proteins/deficiency , Animals , Apoptosis , Brain Stem/pathology , Caspase 3 , Caspases/metabolism , Cell Count , Cell Survival/genetics , Central Nervous System/metabolism , Cerebellum/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Genes, Tumor Suppressor , In Situ Nick-End Labeling , Mice , Mice, Knockout , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/classification , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...