Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Cells ; 10(9)2021 09 18.
Article in English | MEDLINE | ID: mdl-34572114

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is expressed in terminally differentiated cells, where it drives development, morphogenesis, and survival. Temporal and spatial kinase activity is regulated by specific activators of Cdk5, dependent on the cell type and environmental factors. In the kidney, Cdk5 is exclusively expressed in terminally differentiated glomerular epithelial cells called podocytes. In glomerular disease, signaling mechanisms via Cdk5 have been addressed by single or combined conventional knockout of known specific activators of Cdk5. A protective, anti-apoptotic role has been ascribed to Cdk5 but not a developmental phenotype, as in terminally differentiated neurons. The effector kinase itself has never been addressed in animal models of glomerular disease. In the present study, conditional and inducible knockout models of Cdk5 were analyzed to investigate the role of Cdk5 in podocyte development and glomerular disease. While mice with podocyte-specific knockout of Cdk5 had no developmental defects and regular lifespan, loss of Cdk5 in podocytes increased susceptibility to glomerular damage in the nephrotoxic nephritis model. Glomerular damage was associated with reduced anti-apoptotic signals in Cdk5-deficient mice. In summary, Cdk5 acts primarily as master regulator of podocyte survival during glomerular disease and-in contrast to neurons-does not impact on glomerular development or maintenance.


Subject(s)
Apoptosis , Cell Differentiation , Cyclin-Dependent Kinase 5/physiology , Glomerulosclerosis, Focal Segmental/pathology , Podocytes/cytology , Animals , Cells, Cultured , Glomerulosclerosis, Focal Segmental/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Podocytes/metabolism , Signal Transduction
2.
Int J Mol Sci ; 22(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34360858

ABSTRACT

Tauopathies are neurodegenerative diseases characterized by abnormal metabolism of misfolded tau proteins and are progressive. Pathological phosphorylation of tau occurs in the retinal ganglion cells (RGCs) after optic nerve injuries. Cyclin-dependent kinase-5 (Cdk5) causes hyperphosphorylation of tau. To determine the roles played by Cdk5 in retinal degeneration, roscovitine, a Cdk5 inhibitor, was injected intravitreally after optic nerve crush (ONC). The neuroprotective effect of roscovitine was determined by the number of Tuj-1-stained RGCs on day 7. The change in the levels of phosphorylated tau, calpain-1, and cleaved α-fodrin was determined by immunoblots on day 3. The expression of P35/P25, a Cdk5 activator, in the RGCs was determined by immunohistochemistry. The results showed that roscovitine reduced the level of phosphorylated tau by 3.5- to 1.6-fold. Calpain-1 (2.1-fold) and cleaved α-fodrin (1.5-fold) were increased on day 3, suggesting that the calpain signaling pathway was activated. P35/P25 was accumulated in the RGCs that were poorly stained by Tuj-1. Calpain inhibition also reduced the increase in phosphorylated tau. The number of RGCs decreased from 2191 ± 178 (sham) to 1216 ± 122 cells/mm2 on day 7, and roscovitine preserved the level at 1622 ± 130 cells/mm2. We conclude that the calpain-mediated activation of Cdk5 is associated with the pathologic phosphorylation of tau.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Optic Nerve Injuries , Retinal Ganglion Cells , Tauopathies , tau Proteins/metabolism , Animals , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Phosphorylation , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Roscovitine/pharmacology , Tauopathies/metabolism , Tauopathies/pathology
3.
Exp Anim ; 70(1): 22-30, 2021 Feb 06.
Article in English | MEDLINE | ID: mdl-32779618

ABSTRACT

Two members of the CDK5 and ABL enzyme substrate (CABLES) family, CABLES1 and CABLES2, share a highly homologous C-terminus. They interact and associate with cyclin-dependent kinase 3 (CDK3), CDK5, and c-ABL. CABLES1 mediates tumor suppression, regulates cell proliferation, and prevents protein degradation. Although Cables2 is ubiquitously expressed in adult mouse tissues at RNA level, the role of CABLES2 in vivo remains unknown. Here, we generated bicistronic Cables2 knock-in reporter mice that expressed CABLES2 tagged with 3×FLAG and 2A-mediated fluorescent reporter tdTomato. Cables2-3×FLAG-2A-tdTomato (Cables2Tom) mice confirmed the expression of Cables2 in various mouse tissues. Interestingly, high intensity of tdTomato fluorescence was observed in the brain, testis and ovary, especially in the corpus luteum. Furthermore, immunoprecipitation analysis using the brain and testis in Cables2Tom/Tom revealed interaction of CABLES2 with CDK5. Collectively, our new Cables2 knock-in reporter model will enable the comprehensive analysis of in vivo CABLES2 function.


Subject(s)
Cell Cycle Proteins/physiology , Gene Knock-In Techniques/methods , Genes, Reporter/genetics , Models, Animal , Models, Genetic , Animals , Brain/metabolism , Cell Cycle Proteins/genetics , Corpus Luteum/metabolism , Cyclin-Dependent Kinase 5/physiology , Female , Gene Expression , Luminescent Proteins , Male , Mice, Inbred C57BL , Testis/metabolism , Red Fluorescent Protein
4.
Int J Mol Sci ; 23(1)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-35008611

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) has been shown to play a critical role in brain development, learning, memory and neural processing in general. Cdk5 is widely distributed in many neuron types in the central nervous system, while its cell-specific role is largely unknown. Our previous study showed that Cdk5 inhibition restored ocular dominance (OD) plasticity in adulthood. In this study, we specifically knocked down Cdk5 in different types of neurons in the visual cortex and examined OD plasticity by optical imaging of intrinsic signals. Downregulation of Cdk5 in parvalbumin-expressing (PV) inhibitory neurons, but not other neurons, reactivated adult mouse visual cortical plasticity. Cdk5 knockdown in PV neurons reduced the evoked firing rate, which was accompanied by an increment in the threshold current for the generation of a single action potential (AP) and hyperpolarization of the resting membrane potential. Moreover, chemogenetic activation of PV neurons in the visual cortex can attenuate the restoration of OD plasticity by Cdk5 inhibition. Taken together, our results suggest that Cdk5 in PV interneurons may play a role in modulating the excitation and inhibition balance to control the plasticity of the visual cortex.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Dominance, Ocular , Neuronal Plasticity , Neurons/metabolism , Visual Cortex/enzymology , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Gene Knockdown Techniques , Male , Mice , Neurons/physiology , Visual Cortex/physiology
5.
Biochimie ; 168: 277-284, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31786229

ABSTRACT

Changes in brain energy metabolism in diabetes mellitus, including increased insulin resistance and mitochondrial dysfunction, are critically involved in diabetes-related neurodegeneration, and associate with early cognitive impairment as well. The aim of this study is to detect the specific phosphorylated-Thr485- AMP-activated protein kinase (AMPK-α2), regulated by cyclin-dependent kinase 5 (Cdk5) paly the inhibitory functional role of AMPK-α2, Which is maybe the link to the accelerated diabetic brain damage progression. Here, we used GK rats, the type 2 diabetic animal model for in vivo studies and performed In vitro kinase assay, high glucose treatment, -phosphorylated mutation and protein expression in both HEK-293T and HT-22 cell lines. In vitro, the results show that murine wild-type AMPK-α2 was phosphorylated by Cdk5 at a (S/T)PX(K/H/R) phosphorylation consensus sequence, which was associated with decreased AMPK-α2 activity. Surprisingly, mutation of Thr485 to alanine in AMPK-α2 results in the abolished Cdk5 effects, demonstrating that Thr485-phosphorylation is critical to AMPK-α2 inhibition by Cdk5. In addition, these alterations in AMPK-α2-phosphorylation and -activity induced by Cdk5 is specific at Thr485. Furthermore, in GK rats, the increased phosphorylated- Thr 485 of AMPK-α2 results in the decreased AMPK-α2 activity, which is correlated with the apoptosis of neurons in hippocamps. After high glucose treatment, the decreased survival showed in AMPK-α2T485A HT-22 cells compared to AMPK-α2WT. The down-regulated of p-CREB, SNAP25, synaptophysin as well as synapsin-1were shown in both GK rats and HT-22 cell line. Meanwhile, pre-treated with either the specific Cdk5-inhibitor (roscovitine) or the antidiabetic AMPK-α2-inhibitor (metformin) could restore the alterations in neuronal protein expression. Our results suggest that Cdk5-mediated phosphorylated- Thr485 in AMPK-α2 may be involved in the pathogenesis of diabetic brain damage.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclin-Dependent Kinase 5/physiology , Diabetes Mellitus , Hippocampus , Neurons , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Metformin/metabolism , Mice , Neurons/metabolism , Neurons/pathology , Rats , Roscovitine/metabolism
6.
J Alzheimers Dis ; 68(3): 843-855, 2019.
Article in English | MEDLINE | ID: mdl-30856110

ABSTRACT

The cyclin-dependent kinase 5 (CDK5) is known as an exceptional component of the CDK family, due to its characteristic regulatory pathways and its atypical roles in comparison to the classical cyclins. Despite its functional uniqueness, CDK5 shares a great part of its structural similarity with other members of the cyclin-dependent kinase family. After its discovery 26 years ago, a progressive set of cellular functions has been associated with this protein kinase, ranging from neuronal migration, axonal guidance, and synaptic plasticity in diverse stages of brain development, including specific and complex cognitive functions. More than 30 substrates for CDK5 have been found in different cellular pathways. Together with its essential physiological roles, a major discovery was the finding twenty years ago that CDK5 participates in neurodegenerative diseases responsible for tau hyperphosphorylations, and, as a consequence, it becomes a neurotoxic factor. This review focuses on the wide roles of CDK5 in the central nervous system, its implications in neurodegeneration, and provides an integrative insight of its involvement in pain modulation, Alzheimer's disease, and other contexts.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Nervous System Physiological Phenomena , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinases , Humans , Nervous System/metabolism , Pain/metabolism
7.
Diabetes Obes Metab ; 20 Suppl 2: 20-27, 2018 09.
Article in English | MEDLINE | ID: mdl-30230180

ABSTRACT

Efficient and accurate protein translation is essential to producing insulin in pancreatic ß-cells. Transfer RNA (tRNA) is known as the key component of the protein translational machinery. Interestingly, tRNA contains a wide variety of chemical modifications, which are posttranscriptionally catalysed by tRNA modifying enzymes. Recent advances in genome-sequencing technology have unveiled a number of genetic variations that are associated with the development of type 2 diabetes (T2D). Some of these mutations are located in the genes of tRNA modifying enzymes. Using cellular and animal models, it has been showed that dysregulation of tRNA modification impairs protein translation in pancreatic ß-cells and leads to aberrant insulin production. In this review, we discuss the recent findings in the molecular functions of tRNA modifications and their involvement in the development of T2D.


Subject(s)
Insulin-Secreting Cells/physiology , RNA, Transfer/physiology , Alternative Splicing/genetics , Animals , Blood Glucose/metabolism , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/physiology , DNA Methylation/physiology , Diabetes Mellitus, Type 2/genetics , Humans , Insulin/biosynthesis , Insulin Secretion/physiology , Mice, Knockout , Oxidation-Reduction , Phenotype , Polymorphism, Single Nucleotide/physiology , Signal Transduction/physiology , tRNA Methyltransferases/deficiency , tRNA Methyltransferases/genetics , tRNA Methyltransferases/physiology
8.
Exp Neurol ; 306: 92-104, 2018 08.
Article in English | MEDLINE | ID: mdl-29729246

ABSTRACT

Myelination of the central nervous system is important for normal motor and sensory neuronal function and recent studies also link it to efficient learning and memory. Cyclin-dependent kinase 5 (Cdk5) is required for normal oligodendrocyte development, myelination and myelin repair. Here we show that conditional deletion of Cdk5 by targeting with CNP (CNP;Cdk5 CKO) results in hypomyelination and disruption of the structural integrity of Nodes of Ranvier. In addition, CNP;Cdk5 CKO mice exhibited a severe impairment of learning and memory compared to controls that may reflect perturbed neuron-glial interactions. Co-culture of cortical neurons with CNP;Cdk5 CKO oligodendrocyte lineage cells resulted in a significant reduction in the density of neuronal dendritic spines. In short term fear-conditioning studies, CNP;Cdk5 CKO mice had decreased hippocampal levels of immediate early genes such as Arc and Fos, and lower levels of p-CREB and p-cofilin suggested these pathways are affected by the levels of myelination. The novel roles of Cdk5 in oligodendrocyte lineage cells may provide insights for helping understand the cognitive changes sometimes seen in demyelinating diseases such as multiple sclerosis.


Subject(s)
Cyclin-Dependent Kinase 5/genetics , Learning/physiology , Memory/physiology , Oligodendroglia/physiology , Ranvier's Nodes/genetics , Animals , Conditioning, Operant/physiology , Cyclin-Dependent Kinase 5/physiology , Dendritic Spines/physiology , Fear , Female , Gene Deletion , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin Sheath/physiology , Psychomotor Performance/physiology
9.
J Physiol ; 595(1): 363-384, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27461471

ABSTRACT

KEY POINTS: Physical inactivity, which drastically increases with advancing age, is associated with numerous chronic diseases. The nucleus accumbens (the pleasure and reward 'hub' in the brain) influences wheel running behaviour in rodents. RNA-sequencing and subsequent bioinformatics analysis led us to hypothesize a potential relationship between the regulation of dendritic spine density, the molecules involved in synaptic transmission, and age-related reductions in wheel running. Upon completion of follow-up studies, we developed the working model that synaptic plasticity in the nucleus accumbens is central to age-related changes in voluntary running. Testing this hypothesis, inhibition of Cdk5 (comprising a molecule central to the processes described above) in the nucleus accumbens reduced wheel running. The results of the present study show that reductions in synaptic transmission and Cdk5 function are related to decreases in voluntary running behaviour and provide guidance for understanding the neural mechanisms that underlie age-dependent reductions in the motivation to be physically active. ABSTRACT: Increases in age are often associated with reduced levels of physical activity, which, in turn, associates with the development of numerous chronic diseases. We aimed to assess molecular differences in the nucleus accumbens (NAc) (a specific brain nucleus postulated to influence rewarding behaviour) with respect to wheel running and sedentary female Wistar rats at 8 and 14 weeks of age. RNA-sequencing was used to interrogate transcriptomic changes between 8- and 14-week-old wheel running rats, and select transcripts were later analysed by quantitative RT-PCR in age-matched sedentary rats. Voluntary wheel running was greatest at 8 weeks and had significantly decreased by 12 weeks. From 619 differentially expressed mRNAs, bioinformatics suggested that cAMP-mediated signalling, dopamine- and cAMP-regulated neuronal phosphoprotein of 32 kDa feedback, and synaptic plasticity were greater in 8- vs. 14-week-old rats. In depth analysis of these networks showed significant (∼20-30%; P < 0.05) decreases in cell adhesion molecule (Cadm)4 and p39 mRNAs, as well as their proteins from 8 to 14 weeks of age in running and sedentary rats. Furthermore, Cadm4, cyclin-dependent kinase 5 (Cdk5) and p39 mRNAs were significantly correlated with voluntary running distance. Analysis of dendritic spine density in the NAc showed that wheel access increased spine density (P < 0.001), whereas spine density was lower in 14- vs. 8-week-old sedentary rats (P = 0.03). Intriguingly, intra-NAc injection of the Cdk5 inhibitor roscovitine, dose-dependently decreased wheel running. Collectively, these experiments suggest that an age-dependent loss in synaptic function and Cdk5/p39 activity in the NAc may be partially responsible for age-related declines in voluntary running behaviour.


Subject(s)
Aging/physiology , Cyclin-Dependent Kinase 5/physiology , Motivation/physiology , Motor Activity/physiology , Nucleus Accumbens/physiology , Animals , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Female , Neuronal Plasticity/physiology , Purines/pharmacology , Rats, Wistar , Roscovitine , Synaptic Transmission/physiology
10.
Science ; 353(6297): 399-403, 2016 Jul 22.
Article in English | MEDLINE | ID: mdl-27463676

ABSTRACT

Cancers often evade immune surveillance by adopting peripheral tissue- tolerance mechanisms, such as the expression of programmed cell death ligand 1 (PD-L1), the inhibition of which results in potent antitumor immunity. Here, we show that cyclin-dependent kinase 5 (Cdk5), a serine-threonine kinase that is highly active in postmitotic neurons and in many cancers, allows medulloblastoma (MB) to evade immune elimination. Interferon-γ (IFN-γ)-induced PD-L1 up-regulation on MB requires Cdk5, and disruption of Cdk5 expression in a mouse model of MB results in potent CD4(+) T cell-mediated tumor rejection. Loss of Cdk5 results in persistent expression of the PD-L1 transcriptional repressors, the interferon regulatory factors IRF2 and IRF2BP2, which likely leads to reduced PD-L1 expression on tumors. Our finding highlights a central role for Cdk5 in immune checkpoint regulation by tumor cells.


Subject(s)
B7-H1 Antigen/genetics , Cerebellar Neoplasms/immunology , Cyclin-Dependent Kinase 5/physiology , Gene Expression Regulation, Neoplastic , Medulloblastoma/immunology , Neoplasms, Experimental/immunology , Tumor Escape/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cyclin-Dependent Kinase 5/genetics , Humans , Immunologic Surveillance , Interferon Regulatory Factor-2/genetics , Interferon Regulatory Factor-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Oncotarget ; 7(26): 39148-39161, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27283769

ABSTRACT

Cyclin-dependent kinase 5 (CDK5) regulates important neuronal functions via p35. p35 undergoes cleavage in response to neuronal activity and neurotoxic conditions to release its subunit p25. Although p25 has been implicated in various neurodegenerative diseases, the mechanisms by which p25 mediates neurodegenerative impairment have not been fully elucidated. We aimed to determine the role of p25-mediated neurodegeneration on neurogenesis in an inducible transgenic mouse line overexpressing p25 (p25 TG) in the forebrain. Adult neuronal progenitor cells (NPCs) were labeled with BrdU in vivo, which were significantly increased in numbers in the subventricular zone, the hippocampus, and the cortex of p25 TG mice. Consistently, more mitotic cells were observed in p25 TG mice than in controls, even in the cortex and the CA1, which are not neurogenic regions. BrdU-positive cells were negative for GFAP or γ-H2AX, suggesting that they are not astrocytes or dying cells. Neurospheres derived from the dentate gyrus and the cortex were significantly increased in p25 TG mice and can be differentiated into astrocytes and neurons. However, p25 TG decreased the long-term survival of proliferating NPCs and severely impaired adult neurogenesis. A Transwell co-culture system was used to assess the influence of p25-expressing primary neurons on adult NPCs. Co-culture with p25-expressing neurons downregulated Ki67 expression and upregulated cleaved caspase-3, indicating that the paracrine signaling in cell-cell communication is essential for NPC survival and proliferation. Moreover, increased CDK5 activity impairs Wnt activation. This study demonstrates that hyperactivation of p25 may temporarily enhance NPC proliferation, but impair their long-term survival.


Subject(s)
Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/physiology , Neurons/metabolism , Animals , Astrocytes/cytology , Brain Mapping , Cell Proliferation , Coculture Techniques , HEK293 Cells , Hippocampus/metabolism , Humans , Mice , Mice, Transgenic , Mitosis , Neurogenesis , Signal Transduction , Stem Cells/cytology , Treatment Outcome
12.
Cell Death Differ ; 23(2): 333-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26206088

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase and its dysregulation is implicated in neurodegenerative diseases. Likewise, C-terminus of Hsc70-interacting protein (CHIP) is linked to neurological disorders, serving as an E3 ubiquitin ligase for targeting damaged or toxic proteins for proteasomal degradation. Here, we demonstrate that CHIP is a novel substrate for Cdk5. Cdk5 phosphorylates CHIP at Ser20 via direct binding to a highly charged domain of CHIP. Co-immunoprecipitation and ubiquitination assays reveal that Cdk5-mediated phosphorylation disrupts the interaction between CHIP and truncated apoptosis-inducing factor (tAIF) without affecting CHIP's E3 ligase activity, resulting in the inhibition of CHIP-mediated degradation of tAIF. Lentiviral transduction assay shows that knockdown of Cdk5 or overexpression of CHIP(S20A), but not CHIP(WT), attenuates tAIF-mediated neuronal cell death induced by hydrogen peroxide. Thus, we conclude that Cdk5-mediated phosphorylation of CHIP negatively regulates its neuroprotective function, thereby contributing to neuronal cell death progression following neurotoxic stimuli.


Subject(s)
Apoptosis Inducing Factor/physiology , Apoptosis , Cyclin-Dependent Kinase 5/physiology , Neurons/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Cyclin-Dependent Kinase 5/chemistry , Hydrogen Peroxide/pharmacology , Male , Mice, Transgenic , Neurons/drug effects , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
13.
J Neurosci ; 35(45): 15127-34, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26558783

ABSTRACT

The proper growth and arborization of dendrites in response to sensory experience are essential for neural connectivity and information processing in the brain. Although neuronal activity is important for sculpting dendrite morphology, the underlying molecular mechanisms are not well understood. Here, we report that cyclin-dependent kinase 5 (Cdk5)-mediated transcriptional regulation is a key mechanism that controls activity-dependent dendrite development in cultured rat neurons. During membrane depolarization, Cdk5 accumulates in the nucleus to regulate the expression of a subset of genes, including that of the neurotrophin brain-derived neurotrophic factor, for subsequent dendritic growth. Furthermore, Cdk5 function is mediated through the phosphorylation of methyl-CpG-binding protein 2, a key transcriptional repressor that is mutated in the mental disorder Rett syndrome. These findings collectively suggest that the nuclear import of Cdk5 is crucial for activity-dependent dendrite development by regulating neuronal gene transcription during neural development. SIGNIFICANCE STATEMENT: Neural activity directs dendrite development through the regulation of gene transcription. However, how molecular signals link extracellular stimuli to the transcriptional program in the nucleus remains unclear. Here, we demonstrate that neuronal activity stimulates the translocation of the kinase Cdk5 from the cytoplasmic compartment into the nucleus; furthermore, the nuclear localization of Cdk5 is required for dendrite development in cultured neurons. Genome-wide transcriptome analysis shows that Cdk5 deficiency specifically disrupts activity-dependent gene transcription of bdnf. The action of Cdk5 is mediated through the modulation of the transcriptional repressor methyl-CpG-binding protein 2. Therefore, this study elucidates the role of nuclear Cdk5 in the regulation of activity-dependent gene transcription and dendritic growth.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Dendrites/metabolism , Gene Expression Regulation, Developmental , Visual Cortex/growth & development , Visual Cortex/metabolism , Animals , Cells, Cultured , Dendrites/genetics , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Rats
14.
Oncotarget ; 6(26): 21799-801, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26327394

ABSTRACT

Lymphatic vessel dysfunction is associated with various pathologic conditions, including immunologic disorders, lymphedema, as well as tumor dissemination. Yet, the knowledge about the regulation of lymphatic vessel development is still limited. Our study elucidates cyclin dependent kinase 5 (Cdk5) as an essential player in the development of lymphatic vessels. Deletion of Cdk5 in the mouse endothelium results in severe lymphedema formation and embryonic lethality. On the mechanistic level, we show that Cdk5 phosphorylates the forkhead transcription factor Foxc2 which regulates Foxc2-dependent transcription. In summary, our study elucidates the Cdk5-Foxc2 interaction as a critical regulator of lymphatic vessel development.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Forkhead Transcription Factors/metabolism , Lymphangiogenesis/physiology , Lymphatic Vessels/cytology , Lymphatic Vessels/physiology , Animals , Mice , Mice, Knockout
15.
PLoS One ; 10(7): e0131833, 2015.
Article in English | MEDLINE | ID: mdl-26146988

ABSTRACT

Cyclin-dependent kinase 5 (CDK5) is a cytoplasmic serine/ threonine kinase. Knockdown of CDK5 enhances paclitaxel sensitivity in human ovarian cancer cells. This study explores the mechanisms by which CDK5 regulates paclitaxel sensitivity in human ovarian cancers. Multiple ovarian cancer cell lines and xenografts were treated with CDK5 small interfering RNA (siRNA) with or without paclitaxel to examine the effect on cancer cell viability, cell cycle arrest and tumor growth. CDK5 protein was measured by immunohistochemical staining of an ovarian cancer tissue microarray to correlate CDK5 expression with overall patient survival. Knockdown of CDK5 with siRNAs inhibits activation of AKT which significantly correlates with decreased cell growth and enhanced paclitaxel sensitivity in ovarian cancer cell lines. In addition, CDK5 knockdown alone and in combination with paclitaxel induced G1 cell cycle arrest and caspase 3 dependent apoptotic cell death associated with post-translational upregulation and nuclear translocation of TP53 and p27(Kip1) as well as TP53-dependent transcriptional induction of p21(Cip1) in wild type TP53 cancer cells. Treatment of HEYA8 and A2780 wild type TP53 xenografts in nu/nu mice with CDK5 siRNA and paclitaxel produced significantly greater growth inhibition than either treatment alone. Increased expression of CDK5 in human ovarian cancers correlates inversely with overall survival. CDK5 modulates paclitaxel sensitivity by regulating AKT activation, the cell cycle and caspase-dependent apoptosis. CDK5 inhibition can potentiate paclitaxel activity in human ovarian cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cyclin-Dependent Kinase 5/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 5/genetics , Enzyme Activation , Female , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
16.
Curr HIV Res ; 13(1): 43-54, 2015.
Article in English | MEDLINE | ID: mdl-25760044

ABSTRACT

The advent of more effective antiretroviral therapies has reduced the frequency of HIV dementia, however the prevalence of milder HIV associated neurocognitive disorders [HAND] is actually rising. Neurodegenerative mechanisms in HAND might include toxicity by secreted HIV-1 proteins such as Tat, gp120 and Nef that could activate neuro-inflammatory pathways, block autophagy, promote excitotoxicity, oxidative stress, mitochondrial dysfunction and dysregulation of signaling pathways. Recent studies have shown that Tat could interfere with several signal transduction mechanisms involved in cytoskeletal regulation, cell survival and cell cycle re-entry. Among them, Tat has been shown to hyper-activate cyclin-dependent kinase [CDK] 5, a member of the Ser/Thr CDKs involved in cell migration, angiogenesis, neurogenesis and synaptic plasticity. CDK5 is activated by binding to its regulatory subunit, p35 or p39. For this manuscript we review evidence showing that Tat, via calcium dysregulation, promotes calpain-1 cleavage of p35 to p25, which in turn hyper-activates CDK5 resulting in abnormal phosphorylation of downstream targets such as Tau, collapsin response mediator protein-2 [CRMP2], doublecortin [DCX] and MEF2. We also present new data showing that Tat interferes with the trafficking of CDK5 between the nucleus and cytoplasm. This results in prolonged presence of CDK5 in the cytoplasm leading to accumulation of aberrantly phosphorylated cytoplasmic targets [e.g.: Tau, CRMP2, DCX] that impair neuronal function and eventually lead to cell death. Novel therapeutic approaches with compounds that block Tat mediated hyper-activation of CDK5 might be of value in the management of HAND.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Enzyme Activation/physiology , HIV Infections/complications , HIV-1 , Neurocognitive Disorders/etiology , tat Gene Products, Human Immunodeficiency Virus/physiology , Animals , Calcium/metabolism , Cell Death/physiology , Doublecortin Protein , HIV Infections/metabolism , Humans , Mice , Neurocognitive Disorders/metabolism , Neurons/physiology , Phosphorylation
17.
J Neurosci ; 35(6): 2624-35, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673854

ABSTRACT

Deficiency of cyclin-dependent kinase 5 (Cdk5) has been linked to the death of postmitotic cortical neurons during brain development. We now report that, in mouse cortical neurons, Cdk5 is capable of phosphorylating the transcription factor FOXO1 at Ser249 in vitro and in vivo. Cellular stresses resulting from extracellular stimulation by H2O2 or ß-amyloid promote hyperactivation of Cdk5, FOXO1 nuclear export and inhibition of its downstream transcriptional activity. In contrast, a loss of Cdk5 leads to FOXO1 translocation into the nucleus: a shift due to decreased AKT activity but independent of S249 phosphorylation. Nuclear FOXO1 upregulates transcription of the proapoptotic gene, BIM, leading to neuronal death, which can be rescued when endogenous FOXO1 was replaced by the cytoplasmically localized form of FOXO1, FOXO1-S249D. Cytoplasmic, but not nuclear, Cdk5 attenuates neuronal death by inhibiting FOXO1 transcriptional activity and BIM expression. Together, our findings suggest that Cdk5 plays a novel and unexpected role in the degeneration of postmitotic neurons through modulation of the cellular location of FOXO1, which constitutes an alternative pathway through which Cdk5 deficiency leads to neuronal death.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Forkhead Transcription Factors/physiology , Neurons/metabolism , Subcellular Fractions/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 5/genetics , Cytoplasm/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Oncogene Protein v-akt/metabolism , Oncogene Protein v-akt/physiology , Phosphorylation , Serine/metabolism
18.
J Cell Biol ; 207(5): 627-42, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25452387

ABSTRACT

DLC1 is a tumor suppressor protein whose full activity depends on its presence at focal adhesions, its Rho-GTPase activating protein (Rho-GAP) function, and its ability to bind several ligands, including tensin and talin. However, the mechanisms that regulate and coordinate these activities remain poorly understood. Here we identify CDK5, a predominantly cytoplasmic serine/threonine kinase, as an important regulator of DLC1 functions. The CDK5 kinase phosphorylates four serines in DLC1 located N-terminal to the Rho-GAP domain. When not phosphorylated, this N-terminal region functions as an autoinhibitory domain that places DLC1 in a closed, inactive conformation by efficiently binding to the Rho-GAP domain. CDK5 phosphorylation reduces this binding and orchestrates the coordinate activation DLC1, including its localization to focal adhesions, its Rho-GAP activity, and its ability to bind tensin and talin. In cancer, these anti-oncogenic effects of CDK5 can provide selective pressure for the down-regulation of DLC1, which occurs frequently in tumors, and can contribute to the pro-oncogenic activity of CDK5 in lung adenocarcinoma.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , GTPase-Activating Proteins/metabolism , Protein Processing, Post-Translational , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line, Tumor , Focal Adhesions/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
19.
J Biol Chem ; 289(38): 26618-26629, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25100724

ABSTRACT

Thiazolidinedione class of anti-diabetic drugs which are known as peroxisome proliferator-activated receptor γ (PPARγ) ligands have been used to treat metabolic disorders, but thiazolidinediones can also cause several severe side effects, including congestive heart failure, fluid retention, and weight gain. In this study, we describe a novel synthetic PPARγ ligand UNIST HYUNDAI Compound 1 (UHC1) that binds tightly to PPARγ without the classical agonism and which blocks cyclin-dependent kinase 5 (CDK5)-mediated PPARγ phosphorylation. We modified the non-agonist PPARγ ligand SR1664 chemically to improve its solubility and then developed a novel PPARγ ligand, UHC1. According to our docking simulation, UHC1 occupied the ligand-binding site of PPARγ with a higher docking score than SR1664. In addition, UHC1 more potently blocked CDK5-mediated PPARγ phosphorylation at Ser-273. Surprisingly, UHC1 treatment effectively ameliorated the inflammatory response both in vitro and in high-fat diet-fed mice. Furthermore, UHC1 treatment dramatically improved insulin sensitivity in high-fat diet-fed mice without causing fluid retention and weight gain. Taken together, compared with SR1664, UHC1 exhibited greater beneficial effects on glucose and lipid metabolism by blocking CDK5-mediated PPARγ phosphorylation, and these data indicate that UHC1 could be a novel therapeutic agent for use in type 2 diabetes and related metabolic disorders.


Subject(s)
Benzoates/pharmacology , Cyclin-Dependent Kinase 5/physiology , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Indoles/pharmacology , PPAR gamma/metabolism , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Benzoates/chemistry , Binding Sites , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Hydrogen Bonding , Hypoglycemic Agents/chemistry , Indoles/chemistry , Inflammation Mediators/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , PPAR gamma/agonists , PPAR gamma/chemistry , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Rats, Sprague-Dawley
20.
Neurobiol Aging ; 35(12): 2870-2880, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25104559

ABSTRACT

Raf kinase inhibitor protein (RKIP) is a major negative mediator of the extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway. The downregulation of RKIP is correlated with many cancers, but the mechanisms that underlie this downregulation and its roles in the nervous system remain unclear. Here, we demonstrate that RKIP is a substrate of cyclin-dependent kinase 5 (CDK5) in neurons and that the phosphorylation of RKIP at T42 causes the release of Raf-1. Moreover, T42 phosphorylation promotes the exposure and recognition of the target motif "KLYEQ" in the C-terminus of RKIP by chaperone Hsc70 and the subsequent degradation of RKIP via chaperone-mediated autophagy (CMA). Furthermore, in the brain sample of Parkinson's disease (PD) patients and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride-induced and transgenic PD models, we demonstrate that CDK5-mediated phosphorylation and autophagy of RKIP are involved in the overactivation of the ERK/MAPK cascade, leading to S-phase reentry and neuronal loss. These findings provide evidence for the role of the CDK5/RKIP/ERK pathway in PD pathogenesis and suggest that this pathway may be a suitable therapeutic target in PD.


Subject(s)
Autophagy/genetics , Cyclin-Dependent Kinase 5/physiology , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Phosphatidylethanolamine Binding Protein/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , MAP Kinase Signaling System/physiology , Molecular Targeted Therapy , Parkinson Disease/therapy , Phosphatidylethanolamine Binding Protein/physiology , Phosphorylation/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...