Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Nat Neurosci ; 10(1): 67-76, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143272

ABSTRACT

The development of dendritic spines is thought to be crucial for synaptic plasticity. Dendritic spines are retracted upon Eph receptor A4 (EphA4) activation, but the mechanisms that control this process are not well understood. Here we report an important function of cyclin-dependent kinase 5 (Cdk5) in EphA4-dependent spine retraction in mice. We found that blocking Cdk5 activity inhibits ephrin-A1-triggered spine retraction and reduction of mEPSC frequency at hippocampal synapses. The activation of EphA4 resulted in the recruitment of Cdk5 to EphA4, leading to the tyrosine phosphorylation and activation of Cdk5. EphA4 and Cdk5 then enhanced the activation of ephexin1, a guanine-nucleotide exchange factor that regulates activation of the small Rho GTPase RhoA. The association between EphA4 and ephexin1 was significantly reduced in Cdk5(-/-) brains and Cdk5-dependent phosphorylation of ephexin1 was required for the ephrin-A1-mediated regulation of spine density. These findings suggest that ephrin-A1 promotes EphA4-dependent spine retraction through the activation of Cdk5 and ephexin1, which in turn modulates actin cytoskeletal dynamics.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Dendritic Spines/physiology , Guanine Nucleotide Exchange Factors/physiology , Receptor, EphA4/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Dendritic Spines/drug effects , Embryo, Mammalian , Enzyme Activation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Humans , In Vitro Techniques , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/cytology , Patch-Clamp Techniques/methods , Phosphorylation , Receptor, EphA1/genetics , Receptor, EphA1/metabolism , Transfection/methods , Tyrosine/metabolism , rhoA GTP-Binding Protein/metabolism
8.
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
9.
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
10.
J Neurosci ; 28(19): 4872-7, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18463240

ABSTRACT

Accumulating evidence indicates that deregulation of cyclin-dependent kinase 5 (Cdk5) activity is associated with apoptosis in various neurodegenerative disease models. Interestingly, recent studies suggest that Cdk5 may also favor neuronal survival. Nonetheless, whether Cdk5 is directly required for neuronal survival during development remains enigmatic. In the current study, we established the pivotal role of Cdk5 in neuronal survival during development by demonstrating that reduction or absence of Cdk5 activity markedly exacerbated neuronal death in cultures and in vivo. Interestingly, the antiapoptotic protein Bcl-2 (B-cell lymphoma protein 2) was identified as a novel substrate of Cdk5. We found that Cdk5-mediated phosphorylation of Bcl-2 at Ser70 was required for the neuroprotective effect of Bcl-2. Intriguingly, inhibition of this phosphorylation conferred proapoptotic property to Bcl-2. Furthermore, overexpression of a Bcl-2 mutant lacking the Cdk5 phosphorylation site abolished the protective effect of Cdk5 re-expression in Cdk5(-/-) neurons, suggesting that Ser70 phosphorylation of Bcl-2 contributed to Cdk5-mediated neuronal survival. Our observations revealed that Cdk5-mediated Bcl-2 phosphorylation is pivotal for the antiapoptotic effect of Bcl-2 and contributes to the maintenance of neuronal survival by Cdk5. Our study has also identified Cdk5 as a regulator of Bcl-2 function in neuronal apoptosis.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis/physiology , Cell Survival/physiology , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Neuroprotective Agents/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/physiology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Substrate Specificity
11.
J Neurosci ; 28(35): 8747-55, 2008 Aug 27.
Article in English | MEDLINE | ID: mdl-18753376

ABSTRACT

Huntington's disease (HD) is a polyglutamine [poly(Q)] disease with an expanded poly(Q) stretch in the N terminus of the huntingtin protein (htt). A major pathological feature of HD neurons is inclusion bodies, detergent-insoluble aggregates composed of poly(Q)-expanded mutant htt (mhtt). Misfolding of mhtt is thought to confer a toxic property via formation of aggregates. Although toxic molecular species are still debated, it is important to clarify the aggregation mechanism to understand the pathogenesis of mhtt. We show Cdk5/p35 suppresses the formation of mhtt inclusion bodies in cell lines and primary neurons. Although we expressed the N-terminal exon 1 fragment of htt lacking phosphorylation sites for Cdk5 in COS-7 cells, the kinase activity of Cdk5 was required for the suppression. Furthermore, Cdk5/p35 suppressed inclusion formation of atrophin-1, another poly(Q) protein, raising the possibility that Cdk5/p35 generally suppresses inclusion formation of poly(Q) proteins. Microtubules (MTs) were a downstream component of Cdk5/p35 in the suppression of inclusion formation; Cdk5/p35 disrupted MTs, which were required for the formation of inclusions. Moreover, stabilization of MTs by Taxol induced inclusions even with overexpression of Cdk5/p35. The formation of inclusions was also regulated by manipulating the Cdk5/p35 activity in primary rat or mouse cortical neuron cultures. These results indicate that Cdk5-dependent regulation of MT organization is involved in the development of aggregate formation and subsequent pathogenesis of poly(Q) diseases. This Cdk5 inhibition of htt aggregates is a novel mechanism different from htt phosphorylation and interaction with Cdk5 reported previously (Luo et al., 2005; Anne et al., 2007).


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Microtubules/physiology , Mutation , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nuclear Proteins/metabolism , Phosphotransferases/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/deficiency , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Inclusion Bodies/metabolism , Mice , Mice, Knockout , Microtubules/drug effects , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nuclear Proteins/genetics , RNA, Small Interfering/pharmacology , Rats , Transfection/methods , Trinucleotide Repeat Expansion/genetics
12.
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
13.
J Neurosci ; 27(47): 12829-38, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18032654

ABSTRACT

At the lateral wall of the lateral ventricles in the adult rodent brain, neuroblasts form an extensive network of elongated cell aggregates called chains in the subventricular zone and migrate toward the olfactory bulb. The molecular mechanisms regulating this migration of neuroblasts are essentially unknown. Here, we report a novel role for cyclin-dependent kinase 5 (Cdk5), a neuronal protein kinase, in this process. Using in vitro and in vivo conditional knock-out experiments, we found that Cdk5 deletion impaired the chain formation, speed, directionality, and leading process extension of the neuroblasts in a cell-autonomous manner. These findings suggest that Cdk5 plays an important role in neuroblast migration in the postnatal subventricular zone.


Subject(s)
Cell Movement/physiology , Cyclin-Dependent Kinase 5/physiology , Lateral Ventricles/growth & development , Neurons/enzymology , Stem Cells/physiology , Animals , Animals, Newborn , Cell Line , Coculture Techniques , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Humans , Lateral Ventricles/cytology , Lateral Ventricles/enzymology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Neurons/cytology , Stem Cells/enzymology
14.
J Neurosci ; 27(47): 12967-76, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18032670

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) regulates dopamine neurotransmission and has been suggested to serve as a homeostatic target of chronic psychostimulant exposure. To study the role of Cdk5 in the modulation of the cellular and behavioral effects of psychoactive drugs of abuse, we developed Cre/loxP conditional knock-out systems that allow temporal and spatial control of Cdk5 expression in the adult brain. Here, we report the generation of Cdk5 conditional knock-out (cKO) mice using the alphaCaMKII promoter-driven Cre transgenic line (CaMKII-Cre). In this model system, loss of Cdk5 in the adult forebrain increased the psychomotor-activating effects of cocaine. Additionally, these CaMKII-Cre Cdk5 cKO mice show enhanced incentive motivation for food as assessed by instrumental responding on a progressive ratio schedule of reinforcement. Behavioral changes were accompanied by increased excitability of medium spiny neurons in the nucleus accumbens (NAc) in Cdk5 cKO mice. To study NAc-specific effects of Cdk5, another model system was used in which recombinant adeno-associated viruses expressing Cre recombinase caused restricted loss of Cdk5 in NAc neurons. Targeted knock-out of Cdk5 in the NAc facilitated cocaine-induced locomotor sensitization and conditioned place preference for cocaine. These results suggest that Cdk5 acts as a negative regulator of neuronal excitability in the NAc and that Cdk5 may govern the behavioral effects of cocaine and motivation for reinforcement.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/enzymology , Cyclin-Dependent Kinase 5/physiology , Motivation , Neurons/enzymology , Reward , Animals , Corpus Striatum/drug effects , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects
15.
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
16.
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
17.
J Neurosci ; 25(42): 9658-68, 2005 Oct 19.
Article in English | MEDLINE | ID: mdl-16237170

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a serine/threonine kinase with significant homology to cell cycle-related Cdks but is not believed to be active in a typical cell cycle. In Cdk5-deficient embryos and Cdk5 chimeras, migration and survival of postmitotic neurons is compromised in a cell-autonomous manner. In the present study, we show that loss of Cdk5 leads to both failure of neuronal differentiation and loss of cell cycle control. Using specific cytoskeletal proteins as indices of neuronal differentiation, we find that Cdk5-deficient neurons are significantly arrested or delayed in their developmental program both in vivo and in vitro. For example, immunocytochemistry of embryonic day 16 (E16) cortex reveals that the expression of microtubule-associated protein 2c (Map-2c), a marker of mature neurons, is nearly absent in Cdk5(-/-) cells that have migrated to the cortical plate while these same cells continue to express nestin. Similarly, in vitro, Map-2-positive cells are rare in cultures from E16 Cdk5(-/-) embryos. Cell cycle control is also deficient in Cdk5(-/-) cells. In vivo, neurons engaged in cell cycle activities are found in the cortical plate, and, in vitro, class III beta-tubulin-positive cells continue to label with bromodeoxyuridine even after 5 d of incubation. Transfection of a wild-type Cdk5 construct reveals that cell cycle control can be regained in Cdk5(-/-) cells by overexpression of Cdk5. These data indicate that Cdk5 is necessary for both neuronal differentiation and cell cycle inhibition.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Cyclin-Dependent Kinase 5/biosynthesis , Neurons/cytology , Neurons/enzymology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , Embryo, Mammalian , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
18.
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
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL