Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 346: 113866, 2021 12.
Article in English | MEDLINE | ID: mdl-34537209

ABSTRACT

Attention deficit/Hyperactivity disorder (ADHD) is one of the most diagnosed psychiatric disorders nowadays. The core symptoms of the condition include hyperactivity, impulsiveness and inattention. The main pharmacological treatment consists of psychostimulant drugs affecting Dopamine Transporter (DAT) function. We have previously shown that genetically modified mice lacking p35 protein (p35KO), which have reduced Cdk5 activity, present key hallmarks resembling those described in animal models useful for studying ADHD. The p35KO mouse displays spontaneous hyperactivity and shows a calming effect of methylphenidate or amphetamine treatment. Interestingly, dopaminergic neurotransmission is altered in these mice as they have an increased Dopamine (DA) content together with a low DA turnover. This led us to hypothesize that the lack of Cdk5 activity affects DAT expression and/or function in this animal model. In this study, we performed biochemical assays, cell-based approaches, quantitative fluorescence analysis and functional studies that allowed us to demonstrate that p35KO mice exhibit decreased DA uptake and reduced cell surface DAT expression levels in the striatum (STR). These findings are supported by in vitro observations in which the inhibition of Cdk5 activity in N2a cells induced a significant increase in constitutive DAT endocytosis with a concomitant increase in DAT localization to recycling endosomes. Taken together, these data provide evidences regarding the role of Cdk5/p35 in DAT expression and function, thus contributing to the knowledge of DA neurotransmission physiology and also providing therapeutic options for the treatment of DA pathologies such as ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/metabolism , Cyclin-Dependent Kinase 5/deficiency , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/biosynthesis , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Cell Line , Cyclin-Dependent Kinase 5/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Enzyme Activation/physiology , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Int J Radiat Oncol Biol Phys ; 108(3): 758-769, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32407930

ABSTRACT

PURPOSE: Tumor resistance to radiation therapy is a therapeutic challenge in the treatment of patients with non-small cell lung cancer. Cyclin-dependent kinase 5 (CDK5) has been proposed to participate in cell proliferation, migration and invasion, drug resistance, and immune evasion. However, the functions and regulatory mechanisms of CDK5 in lung cancer radioresistance have not been investigated. METHODS AND MATERIALS: DNA damage response and repair were measured by neutral comet assay and γ-H2AX and Rad51 foci staining. The biological functions of CDK5 in lung cancer radioresistance were investigated with clonogenic survival assays and xenograft tumor models. Small interfering RNAs and short hairpin RNAs were used to knock down CDK5 in A549 and H1299 cells. The effects of CDK5 depletion on the tumorigenic behaviors of lung cancer cells were evaluated in vitro and in vivo. Gene expression was examined by RNA-seq and quantitative real-time polymerase chain reaction. RESULTS: We report that CDK5 depletion impairs lung cancer progression and radioresistance in vitro and in vivo. Mechanistically, we identify TAZ, a component of the Hippo pathway, as a critical downstream effector of CDK5. Loss of CDK5 downregulates TAZ expression and attenuates Hippo signaling activation. Importantly, we provide evidence that TAZ is the major effector mediating the biological functions of CDK5 in lung cancer. CONCLUSIONS: These results illustrate that CDK5 activates Hippo signaling via TAZ to participate in tumorigenesis and radioresistance, suggesting that CDK5 may be a promising radiosensitization target for the treatment of lung cancer.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Lung Neoplasms/radiotherapy , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/physiology , Transcription Factors/metabolism , A549 Cells , Acyltransferases , Animals , Cell Line, Tumor , Comet Assay , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , DNA Damage , DNA Repair , Disease Progression , Down-Regulation , Fluorescent Antibody Technique , Gene Knockdown Techniques , Gene Silencing , Heterografts , Hippo Signaling Pathway , Histones/analysis , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering , Rad51 Recombinase/analysis , Up-Regulation
3.
Channels (Austin) ; 12(1): 65-75, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29308980

ABSTRACT

Transient receptor potential cation channel, subfamily A, member 1 (TRPA1), is activated by a broad range of noxious stimuli. Cdk5, a member of the Cdk family, has recently been identified as a modulator of pain signaling pathways. In the current study, we investigated the extent to which Cdk5 modulates TRPA1 activity. Cdk5 inhibition was found to attenuate TRPA1 response to agonist in mouse DRG sensory neurons. Additionally, the presence of active Cdk5 was associated with increased TRPA1 phosphorylation in transfected HEK293 cells that was roscovitine-sensitive and absent in the mouse mutant S449A full-length channel. Immunopurified Cdk5 was observed to phosphorylate human TRPA1 peptide substrate at S448A in vitro. Our results point to a role for Cdk5 in modulating TRPA1 activity.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Sensory Receptor Cells/metabolism , TRPA1 Cation Channel/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/deficiency , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Sensory Receptor Cells/drug effects , TRPA1 Cation Channel/antagonists & inhibitors
4.
Mol Neurobiol ; 54(9): 6681-6696, 2017 11.
Article in English | MEDLINE | ID: mdl-27744570

ABSTRACT

Cerebral ischemia is a cerebrovascular episode that generates a high incidence of death and physical and mental disabilities worldwide. Excitotoxicity, release of free radicals, and exacerbated immune response cause serious complications in motor and cognitive areas during both short and long time frames post-ischemia. CDK5 is a kinase that is widely involved in the functions of neurons and astrocytes, and its over-activation is implicated in neurodegenerative processes. In this study, we evaluated the brain parenchymal response to the transplantation of CDK5-knockdown astrocytes into the somatosensory cortex after ischemia in rats. Male Wistar rats were subjected to the two-vessel occlusion (2VO) model of global cerebral ischemia and immediately transplanted with shCDK5miR- or shSCRmiR-transduced astrocytes or with untransduced astrocytes (Control). Our findings showed that animals transplanted with shCDK5miR astrocytes recovered motor and neurological performance better than with those transplanted with WT or shSCRmiR astrocytes. Cell transplantation produced an overall prevention of neuronal loss, and CDK5-knockdown astrocytes significantly increased the immunoreactivity (IR) of endogenous GFAP in branches surrounding blood vessels, accompanied by the upregulation of PECAM-1 IR in the walls of vessels in the motor and somatosensory regions and by an increase in Ki67 IR in the subventricular zone (SVZ), partially associated with the production of BDNF. Together, our data suggest that transplantation of shCDK5miR astrocytes protects the neurovascular unit in ischemic rats, allowing the motor and neurological function recovery.


Subject(s)
Astrocytes/metabolism , Astrocytes/transplantation , Brain Ischemia/metabolism , Brain Ischemia/therapy , Cyclin-Dependent Kinase 5/deficiency , Neuroprotection/physiology , Animals , Cells, Cultured , Gene Knockdown Techniques/methods , Male , Rats , Rats, Wistar
5.
Mol Immunol ; 67(2 Pt B): 317-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26198700

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is known as a unique member of the cyclin-dependent family of serine/threonine kinases. Previously, we demonstrated Cdk5 to be an important regulator of T cell function and that disruption of Cdk5 expression ameliorates T cell mediated neuroinflammation. Here, we show a novel role of Cdk5 in the regulation of Foxp3 expression in murine CD4(+) T cells. Our data indicate that disruption of Cdk5 activity in T cells abrogates the IL-6 suppression of Foxp3 expression. This effect is achieved through Cdk5 phosphorylation of the signal transducer and activator of transcription 3 (Stat3) specifically at Serine 727 in T cells, and we show this post-translational modification is required for proper Stat3 DNA binding to the Foxp3 gene on the enhancer II region. Taken together, our data point to an essential role for Cdk5 in the differentiation of T cells as it regulates Foxp3 gene expression through phosphorylation of Stat3.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Phosphoserine/metabolism , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Cyclin-Dependent Kinase 5/deficiency , Enhancer Elements, Genetic/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Interleukin-6/pharmacology , Mice, Inbred C57BL , Phosphorylation/drug effects , Protein Binding/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/pharmacology
6.
Cell Cycle ; 14(8): 1327-36, 2015.
Article in English | MEDLINE | ID: mdl-25785643

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a unique member of a family of serine/threonine cyclin-dependent protein kinases. We previously demonstrated disruption of Cdk5 gene expression in mice impairs T-cell function and ameliorates T-cell-mediated neuroinflammation. Here, we show Cdk5 modulates gene expression during T-cell activation by impairing the repression of gene transcription by histone deacetylase 1 (HDAC1) through specific phosphorylation of the mSin3a protein at serine residue 861. Disruption of Cdk5 activity in T-cells enhances HDAC activity and binding of the HDAC1/mSin3a complex to the IL-2 promoter, leading to suppression of IL-2 gene expression. These data point to essential roles for Cdk5 in regulating gene expression in T-cells and transcriptional regulation by the co-repressor mSin3a.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Histone Deacetylase 1/metabolism , Interleukin-2/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Gene Expression Regulation/drug effects , Histone Deacetylase 1/genetics , Humans , Interleukin-2/genetics , Jurkat Cells , Leupeptins/pharmacology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sin3 Histone Deacetylase and Corepressor Complex , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Nature ; 517(7534): 391-5, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25409143

ABSTRACT

Obesity-linked insulin resistance is a major precursor to the development of type 2 diabetes. Previous work has shown that phosphorylation of PPARγ (peroxisome proliferator-activated receptor γ) at serine 273 by cyclin-dependent kinase 5 (Cdk5) stimulates diabetogenic gene expression in adipose tissues. Inhibition of this modification is a key therapeutic mechanism for anti-diabetic drugs that bind PPARγ, such as the thiazolidinediones and PPARγ partial agonists or non-agonists. For a better understanding of the importance of this obesity-linked PPARγ phosphorylation, we created mice that ablated Cdk5 specifically in adipose tissues. These mice have both a paradoxical increase in PPARγ phosphorylation at serine 273 and worsened insulin resistance. Unbiased proteomic studies show that extracellular signal-regulated kinase (ERK) kinases are activated in these knockout animals. Here we show that ERK directly phosphorylates serine 273 of PPARγ in a robust manner and that Cdk5 suppresses ERKs through direct action on a novel site in MAP kinase/ERK kinase (MEK). Importantly, pharmacological inhibition of MEK and ERK markedly improves insulin resistance in both obese wild-type and ob/ob mice, and also completely reverses the deleterious effects of the Cdk5 ablation. These data show that an ERK/Cdk5 axis controls PPARγ function and suggest that MEK/ERK inhibitors may hold promise for the treatment of type 2 diabetes.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Diabetes Mellitus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , PPAR gamma/metabolism , Adipocytes/enzymology , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Diet, High-Fat , Humans , Insulin Resistance , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Obese , PPAR gamma/chemistry , Phosphorylation
8.
Mol Brain ; 7: 82, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25404232

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 5 (Cdk5), which is activated by binding to p35 or p39, is involved in synaptic plasticity and affects learning and memory formation. In Cdk5 knockout (KO) mice and p35 KO mice, brain development is severely impaired because neuronal migration is impaired and lamination is disrupted. To avoid these developmental confounders, we generated inducible CreER-p35 conditional (cKO) mice to study the role of Cdk5/p35 in higher brain function. RESULTS: CreER-p35 cKO mice exhibited spatial learning and memory impairments and reduced anxiety-like behavior. These phenotypes resulted from a decrease in the dendritic spine density of CA1 pyramidal neurons and defective long-term depression induction in the hippocampus. CONCLUSIONS: Taken together, our findings reveal that Cdk5/p35 regulates spatial learning and memory, implicating Cdk5/p35 as a therapeutic target in neurological disorders.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Memory , Phosphotransferases/metabolism , Spatial Learning , Administration, Oral , Animals , Anxiety/metabolism , Anxiety/physiopathology , Cyclin-Dependent Kinase 5/deficiency , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Integrases/metabolism , Long-Term Synaptic Depression/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Phosphorylation/drug effects , Phosphotransferases/deficiency , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, Estrogen/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Tamoxifen/administration & dosage , Tamoxifen/pharmacology
9.
J Neurosci ; 34(31): 10415-29, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080600

ABSTRACT

Failure of remyelination in diseases, such as multiple sclerosis (MS), leads to permanent axonal damage and irreversible functional loss. The mechanisms controlling remyelination are currently poorly understood. Recent studies implicate the cyclin-dependent kinase 5 (Cdk5) in regulating oligodendrocyte (OL) development and myelination in CNS. In this study, we show that Cdk5 is also an important regulator of remyelination. Pharmacological inhibition of Cdk5 inhibits repair of lysolecithin lesions. This inhibition is a consequence of Cdk5 disruption in neural cells because remyelination in slice cultures is blocked by Cdk5 inhibitors, whereas specific deletion of Cdk5 in OLs inhibits myelin repair. In CNP-Cre;Cdk5(fl/fl) conditional knock-out mouse (Cdk5 cKO), myelin repair was delayed significantly in response to focal demyelinating lesions compared with wild-type animals. The lack of myelin repair was reflected in decreased expression of MBP and proteolipid protein and a reduction in the total number of myelinated axons in the lesion. The number of CC1(+) cells in the lesion sites was significantly reduced in Cdk5 cKO compared with wild-type animals although the total number of oligodendrocyte lineage cells (Olig2(+) cells) was increased, suggesting that Cdk5 loss perturbs the transition of early OL lineage cell into mature OL and subsequent remyelination. The failure of remyelination in Cdk5 cKO animals was associated with a reduction in signaling through the Akt pathway and an enhancement of Gsk-3ß signaling pathways. Together, these data suggest that Cdk5 is critical in regulating the transition of adult oligodendrocyte precursor cells to mature OLs that is essential for myelin repair in adult CNS.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Demyelinating Diseases/metabolism , Glycogen Synthase Kinase 3/metabolism , Myelin Sheath/physiology , Oligodendroglia/physiology , Signal Transduction/physiology , Spinal Cord/pathology , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/deficiency , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , Analysis of Variance , Animals , Cerebellum , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Glycogen Synthase Kinase 3 beta , In Vitro Techniques , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Myelin Sheath/ultrastructure , Oligodendroglia/ultrastructure , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord/ultrastructure
10.
PLoS One ; 9(3): e89310, 2014.
Article in English | MEDLINE | ID: mdl-24662752

ABSTRACT

Cyclin dependent kinase 5 (Cdk5) is a proline-directed Ser/Thr kinase involved in various biological functions during normal brain development and neurodegeneration. In brain, Cdk5 activity is specific to post-mitotic neurons, due to neuronal specific expression of its activator p35. The biological functions of Cdk5 have been ascribed to its cytoplasmic substrates, however not much is known in nucleus. Here, we show that nuclear transcription factor Sox6 is a direct nuclear target of Cdk5. Sox6 is expressed in Tuj1 positive neurons, suggesting that Sox6 is expressed in differentiating neurons. The expression of Sox6 is high in mitotic nuclei during embryonic day 12 (E12) and gradually decreases during development into adult. On the other hand, Cdk5 expression gradually increases during its development. We show that Sox6 is expressed in mitotic nuclei in embryonic day 12 (E12) and in migrating neurons of E16. Sox6 is phosphorylated in vivo. Sox6 was detected by phospho-Ser/Thr and phospho-Ser/Thr-Pro and MPM-2 (Mitotic protein #2) antibodies in brain. Furthermore, calf intestinal alkaline phosphatase (CIAP) digestion resulted in faster migration of Sox6 band. The GST-Sox6 was phosphorylated by Cdk5/p35. The mass spectrometry analysis revealed that Sox6 is phosphorylated at T119PER motif. We show that Sox6 steady state levels are regulated by Cdk5. Cdk5 knockout mice die in utero and Sox6 protein expression is remarkably high in Cdk5-/- brain, however, there is no change in mRNA expression, suggesting a post-translational regulation of Sox6 by Cdk5. Transfection of primary cortical neurons with WT Cdk5 reduced Sox6 levels, while dominant negative (DN) Cdk5 and p35 increased Sox6 levels. Thus, our results indicate that Cdk5 regulates Sox6 steady state protein level that has an important role in brain development and function.


Subject(s)
Brain/metabolism , Cyclin-Dependent Kinase 5/metabolism , SOXD Transcription Factors/metabolism , Amino Acid Sequence , Animals , Brain/cytology , Brain/embryology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Knockout Techniques , Humans , Mice , Mitosis/drug effects , Molecular Sequence Data , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphotransferases/metabolism , Purines/pharmacology , Rats , Roscovitine , SOXD Transcription Factors/chemistry , SOXD Transcription Factors/genetics
11.
J Neuroinflammation ; 11: 28, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24495352

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 5 (Cdk5) is essential for brain development and function, and its deregulated expression is implicated in some of neurodegenerative diseases. We reported earlier that the forebrain-specific Cdk5 conditional knockout (cKO) mice displayed an early lethality associated with neuroinflammation, increased expression of the neuronal tissue-type plasminogen activator (tPA), and neuronal migration defects. METHODS: In order to suppress neuroinflammation in the cKO mice, we first treated these mice with pioglitazone, a PPARγ agonist, and analyzed its effects on neuronal loss and longevity. In a second approach, to delineate the precise role of tPA in neuroinflammation in these mice, we generated Cdk5 cKO; tPA double knockout (dKO) mice. RESULTS: We found that pioglitazone treatment significantly reduced astrogliosis, microgliosis, neuronal loss and behavioral deficit in Cdk5 cKO mice. Interestingly, the dKO mice displayed a partial reversal in astrogliosis, but they still died at early age, suggesting that the increased expression of tPA in the cKO mice does not contribute significantly to the pathological process leading to neuroinflammation, neuronal loss and early lethality. CONCLUSION: The suppression of neuroinflammation in Cdk5 cKO mice ameliorates gliosis and neuronal loss, thus suggesting the potential beneficial effects of the PPARγ agonist pioglitazone for the treatment for neurodegenerative diseases.


Subject(s)
Cyclin-Dependent Kinase 5/deficiency , Encephalitis/drug therapy , Encephalitis/mortality , Encephalitis/pathology , PPAR gamma/agonists , Prosencephalon/drug effects , Thiazolidinediones , Animals , Apoptosis/drug effects , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Cytokines/metabolism , Disease Models, Animal , Encephalitis/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gliosis/etiology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , PPAR gamma/metabolism , Phosphotransferases/genetics , Phosphotransferases/metabolism , Pioglitazone , Prosencephalon/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
12.
Nat Neurosci ; 15(11): 1506-15, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064382

ABSTRACT

The neurotrophin brain-derived neurotrophic factor (BDNF) and its receptor TrkB participate in diverse neuronal functions, including activity-dependent synaptic plasticity that is crucial for learning and memory. On binding to BDNF, TrkB is not only autophosphorylated at tyrosine residues but also undergoes serine phosphorylation at S478 by the serine/threonine kinase cyclin-dependent kinase 5 (Cdk5). However, the in vivo function of this serine phosphorylation remains unknown. We generated knock-in mice lacking this serine phosphorylation (Trkb(S478A/S478A) mice) and found that the TrkB phosphorylation-deficient mice displayed impaired spatial memory and compromised hippocampal long-term potentiation (LTP). S478 phosphorylation of TrkB regulates its interaction with the Rac1-specific guanine nucleotide exchange factor TIAM1, leading to activation of Rac1 and phosphorylation of S6 ribosomal protein during activity-dependent dendritic spine remodeling. These findings reveal the importance of Cdk5-mediated S478 phosphorylation of TrkB in activity-dependent structural plasticity, which is crucial for LTP and spatial memory formation.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Long-Term Potentiation/physiology , Memory/physiology , Receptor, trkB/metabolism , Spatial Behavior/physiology , Analysis of Variance , Animals , Brain/cytology , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Disks Large Homolog 4 Protein , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Glutamic Acid/pharmacology , Green Fluorescent Proteins/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanylate Kinases/metabolism , Humans , Immunoprecipitation/methods , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Phosphorylation/genetics , Quinoxalines/pharmacology , Rats , Receptor, trkB/genetics , Ribosomal Protein S6 Kinases , Serine/metabolism , Silver Staining , Synaptophysin/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Time Factors , Transfection , rac1 GTP-Binding Protein/metabolism
13.
Nat Cell Biol ; 14(4): 409-15, 2012 Mar 04.
Article in English | MEDLINE | ID: mdl-22388889

ABSTRACT

Chronic stress in the endoplasmic reticulum (ER) underlies many degenerative and metabolic diseases involving apoptosis of vital cells. A well-established example is autosomal dominant retinitis pigmentosa (ADRP), an age-related retinal degenerative disease caused by mutant rhodopsins. Similar mutant alleles of Drosophila Rhodopsin-1 also impose stress on the ER and cause age-related retinal degeneration in that organism. Well-characterized signalling responses to ER stress, referred to as the unfolded protein response (UPR), induce various ER quality control genes that can suppress such retinal degeneration. However, how cells activate cell death programs after chronic ER stress remains poorly understood. Here, we report the identification of a signalling pathway mediated by cdk5 and mekk1 required for ER-stress-induced apoptosis. Inactivation of these genes specifically suppressed apoptosis, without affecting other protective branches of the UPR. CDK5 phosphorylates MEKK1, and together, they activate the JNK pathway for apoptosis. Moreover, disruption of this pathway can delay the course of age-related retinal degeneration in a Drosophila model of ADRP. These findings establish a previously unrecognized branch of ER-stress response signalling involved in degenerative diseases.


Subject(s)
Apoptosis , Chromosome Aberrations , Cyclin-Dependent Kinase 5/metabolism , Drosophila Proteins/metabolism , Endoplasmic Reticulum Stress , MAP Kinase Kinase Kinase 1/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Amino Acid Sequence , Animals , Apoptosis/genetics , Cell Line , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Disease Models, Animal , Drosophila/metabolism , Drosophila Proteins/deficiency , Genes, Dominant , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Signaling System , Molecular Sequence Data , Retinitis Pigmentosa/metabolism , Rhodopsin/metabolism , Signal Transduction/genetics , Time Factors
14.
Cell Cycle ; 11(8): 1603-10, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22456337

ABSTRACT

Recent studies have shown the involvement of cyclin-dependent kinase 5 (Cdk5) in cell cycle regulation in postmitotic neurons. In this study, we demonstrate that Cdk5 and its co-activator p35 were detected in the nuclear fraction in neurons and Cdk5/p35 phosphorylated retinoblastoma (Rb) protein, a key protein controlling cell cycle re-entry. Cdk5/p35 phosphorylates Rb at the sites similar to those phosphorylated by Cdk4 and Cdk2. Furthermore, increased Cdk5 activity elevates activity of E2F transcription factor, which can trigger cell cycle re-entry, leading to neuronal cell death. A normal Cdk5 activity in neurons did not induce E2F activation, suggesting that Cdk5 does not induce cell cycle re-entry under normal conditions. Taken together, these results indicate that Cdk5 can regulate cell cycle by its ability to phosphorylate Rb. Most importantly, increased Cdk5 activity induces cell cycle re-entry, which is especially detrimental for survival of postmitotic neurons.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , E2F Transcription Factors/metabolism , Neurons/metabolism , Retinoblastoma Protein/metabolism , Amino Acid Sequence , Animals , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Mass Spectrometry , Mice , Molecular Sequence Data , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphopeptides/analysis , Phosphorylation
15.
J Neurosci ; 31(32): 11547-52, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21832185

ABSTRACT

Nestin is expressed in many different progenitors during development including those of the CNS, heart, skeletal muscle, and kidney. The adult expression is mainly restricted to the subependymal zone and dentate gyrus of the brain, the neuromuscular junction, and renal podocytes. In addition, this intermediate filament protein has served as a marker of neural stem/progenitor cells for close to 20 years. Therefore it is surprising that its function in development and adult physiology is still poorly understood. Here we report that nestin deficiency is compatible with normal development of the CNS. The mutant mice, however, show impaired motor coordination. Furthermore, we found that the number of acetylcholine receptor clusters, the nerve length, and the endplate bandwidth are significantly increased in neuromuscular junction area of nestin-deficient mice. This is similar to the phenotype described for deficiency of cyclin-dependent kinase 5 (Cdk5), a candidate downstream affecter of nestin. Moreover, we demonstrate that nestin deficiency can rescue maintenance of acetylcholine receptor clusters in the absence of agrin, similar to Cdk5/agrin double knock-outs, suggesting that the observed nestin deficiency phenotype is the consequence of aberrant Cdk5 activity.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Cyclin-Dependent Kinase 5/deficiency , Intermediate Filament Proteins/deficiency , Nerve Tissue Proteins/deficiency , Neuromuscular Junction/metabolism , Receptor Aggregation/physiology , Receptors, Cholinergic/metabolism , Agrin/deficiency , Agrin/genetics , Agrin/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/physiology , Female , Gene Targeting/methods , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/physiology , Male , Mice , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Nestin , Neuromuscular Junction/physiology , Receptor Aggregation/genetics , Receptors, Cholinergic/genetics , Receptors, Cholinergic/physiology
16.
Mol Biol Cell ; 21(20): 3601-14, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20810788

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) plays a key role in the development of the mammalian nervous system; it phosphorylates a number of targeted proteins involved in neuronal migration during development to synaptic activity in the mature nervous system. Its role in the initial stages of neuronal commitment and differentiation of neural stem cells (NSCs), however, is poorly understood. In this study, we show that Cdk5 phosphorylation of p27(Kip1) at Thr187 is crucial to neural differentiation because 1) neurogenesis is specifically suppressed by transfection of p27(Kip1) siRNA into Cdk5(+/+) NSCs; 2) reduced neuronal differentiation in Cdk5(-/-) compared with Cdk5(+/+) NSCs; 3) Cdk5(+/+) NSCs, whose differentiation is inhibited by a nonphosphorylatable mutant, p27/Thr187A, are rescued by cotransfection of a phosphorylation-mimicking mutant, p27/Thr187D; and 4) transfection of mutant p27(Kip1) (p27/187A) into Cdk5(+/+) NSCs inhibits differentiation. These data suggest that Cdk5 regulates the neural differentiation of NSCs by phosphorylation of p27(Kip1) at theThr187 site. Additional experiments exploring the role of Ser10 phosphorylation by Cdk5 suggest that together with Thr187 phosphorylation, Ser10 phosphorylation by Cdk5 promotes neurite outgrowth as neurons differentiate. Cdk5 phosphorylation of p27(Kip1), a modular molecule, may regulate the progress of neuronal differentiation from cell cycle arrest through differentiation, neurite outgrowth, and migration.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Phosphothreonine/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Proliferation , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Mice , Molecular Sequence Data , Mutation/genetics , Neurites/metabolism , Neurogenesis , Phosphorylation , Phosphoserine/metabolism , Protein Transport , RNA, Small Interfering/metabolism , Substrate Specificity , Transfection
17.
J Neurosci ; 30(15): 5219-28, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20392944

ABSTRACT

Neurons that reenter a cell cycle after maturation are at increased risk for death, yet the mechanisms by which a normal neuron suppresses the cycle remain mostly unknown. Our laboratory has shown that cyclin-dependent kinase 5 (Cdk5) is a potent cell cycle suppressor, and we report here on the molecular basis of this activity. Cell cycle suppression by Cdk5 requires its binding to the p35 activator protein. The related p39 and p25 proteins cannot serve as substitutes. Unexpectedly, Cdk5 enzymatic activity is not required to perform this function. Rather, the link to cell cycle regulation is made through the formation of a previously unknown complex consisting of the p35-Cdk5 dimer and E2F1. Formation of this complex excludes the E2F1 cofactor, DP1, thus inhibiting E2F1 binding to the promoters of various cell cycle genes. This anti-cell cycle activity is most likely a neuroprotective function of Cdk5.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase 5/metabolism , E2F1 Transcription Factor/metabolism , Neurons/physiology , Phosphotransferases/metabolism , Transcription Factor DP1/metabolism , Animals , Brain/enzymology , Brain/physiology , Cell Line, Tumor , Cell Nucleus/physiology , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Cytoplasm/physiology , DNA/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Nerve Tissue Proteins/metabolism , Neurons/enzymology
18.
Am J Pathol ; 176(1): 320-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19948833

ABSTRACT

Neuronal migration disorders are often identified in patients with epilepsy refractory to medical treatment. The prolonged or repeated seizures are known to cause neuronal death; however, the mechanism underlying seizure-induced neuronal death remains to be elucidated. An essential role of cyclin-dependent kinase 5 (Cdk5) in brain development has been demonstrated in Cdk5(-/-) mice, which show neuronal migration defects and perinatal lethality. Here, we show the consequences of Cdk5 deficiency in the postnatal brain by generating Cdk5 conditional knockout mice, in which Cdk5is selectively eliminated from neurons in the developing forebrain. The conditional mutant mice were viable, but exhibited complex neurological deficits including seizures, tremors, and growth retardation. The forebrain not only showed disruption of layering, but also neurodegenerative changes accompanied by neuronal loss and microglial activation. The neurodegenerative changes progressed with age and were accompanied by up-regulation of the neuronal tissue-type plasminogen activator, a serine protease known to mediate microglial activation. Thus age-dependent neurodegeneration in the Cdk5 conditional knockout mouse brain invoked a massive inflammatory reaction. These findings indicate an important role of Cdk5 in inflammation, and also provide a mouse model to examine the possible involvement of inflammation in the pathogenesis of progressive cognitive decline in patients with neuronal migration disorders.


Subject(s)
Cyclin-Dependent Kinase 5/deficiency , Gene Deletion , Microglia/pathology , Nerve Degeneration/enzymology , Neurons/enzymology , Prosencephalon/embryology , Prosencephalon/enzymology , Animals , Cyclin-Dependent Kinase 5/metabolism , Mice , Mice, Knockout , Microglia/enzymology , Nerve Degeneration/pathology , Neurons/pathology , Organ Specificity , Prosencephalon/pathology , Survival Analysis , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
20.
Endocrinology ; 150(1): 396-403, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18755796

ABSTRACT

The roles of cyclin-dependent kinase 5 (Cdk5) in central nervous system and neurodegenerative diseases have been intensely investigated in recent decades. Because protein expressions of Cdk5 and its regulator, p35, have been identified in Leydig cells, it is informative to further explore the novel function of Cdk5/p35 in male reproduction. Here we show that Cdk5/p35 protein expression and kinase activity in mouse Leydig cells are regulated by human chorionic gonadotrophin (hCG) in both dose- and time-dependent manners. Blocking of Cdk5 by molecular inhibitors or small interfering RNA resulted in reduction of testosterone production by Leydig cells. cAMP, a second messenger in LH signaling, was identified as a factor in hCG-dependent regulation of Cdk5/p35. Importantly, Cdk5 protein and kinase activity could support accumulation of steroidogenic acute regulatory (StAR) protein, a crucial component of steroidogenesis. We additionally addressed the protein interaction between Cdk5/p35 and StAR. The Cdk5-dependent serine phosphorylation of StAR indicated a possible mechanism by which Cdk5 induced accumulation of StAR protein. In conclusion, Cdk5 modulates hCG-induced androgen production in mouse Leydig cells, possibly through regulation of StAR protein levels. These results indicate that Cdk5 may play an important role in male reproductive endocrinology and is a potential therapeutic target in androgen-related diseases.


Subject(s)
Androgens/biosynthesis , Cyclin-Dependent Kinase 5/metabolism , Leydig Cells/metabolism , Membrane Transport Proteins/biosynthesis , Phosphoproteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Chorionic Gonadotropin/pharmacology , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Kinetics , Leydig Cells/drug effects , Male , Mice , Phosphoproteins/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...