Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Intervalo de año de publicación
1.
Front Mol Neurosci ; 15: 848642, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35401105

RESUMEN

Disruption of the inhibitory control provided by the glycinergic system is one of the major mechanisms underlying chronic pain. In line with this concept, recent studies have provided robust proof that pharmacological intervention of glycine receptors (GlyRs) restores the inhibitory function and exerts anti-nociceptive effects on preclinical models of chronic pain. A targeted regulation of the glycinergic system requires the identification of the GlyR subtypes involved in chronic pain states. Nevertheless, the roles of individual GlyR subunits in nociception and in chronic pain are yet not well defined. This review aims to provide a systematic outline on the contribution of GlyR subtypes in chronic pain mechanisms, with a particular focus on molecular pathways of spinal glycinergic dis-inhibition mediated by post-translational modifications at the receptor level. The current experimental evidence has shown that phosphorylation of synaptic α1ß and α3ß GlyRs are involved in processes of spinal glycinergic dis-inhibition triggered by chronic inflammatory pain. On the other hand, the participation of α2-containing GlyRs and of ß subunits in pain signaling have been less studied and remain undefined. Although many questions in the field are still unresolved, future progress in GlyR research may soon open new exciting avenues into understanding and controlling chronic pain.

2.
Curr Protoc ; 1(10): e276, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34679246

RESUMEN

Cyclin-dependent kinases (Cdks) are generally known to be involved in controlling the cell cycle, but Cdk5 is a unique member of this protein family for being most active in post-mitotic neurons. Cdk5 is developmentally important in regulating neuronal migration, neurite outgrowth, and axon guidance. Cdk5 is enriched in synaptic membranes and is known to modulate synaptic activity. Postnatally, Cdk5 can also affect neuronal processes such as dopaminergic signaling and pain sensitivity. Dysregulated Cdk5, in contrast, has been linked to neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Despite primarily being implicated in neuronal development and activity, Cdk5 has lately been linked to non-neuronal functions including cancer cell growth, immune responses, and diabetes. Since Cdk5 activity is tightly regulated, a method for measuring its kinase activity is needed to fully understand the precise role of Cdk5 in developmental and disease processes. This article includes methods for detecting Cdk5 kinase activity in cultured cells or tissues, identifying new substrates, and screening for new kinase inhibitors. Furthermore, since Cdk5 shares homology and substrate specificity with Cdk1 and Cdk2, the Cdk5 kinase assay can be used, with modification, to measure the activity of other Cdks as well. © 2021 Wiley Periodicals LLC. This article has been contributed to by US Government employees and their work is in the public domain in the USA. Basic Protocol 1: Measuring Cdk5 activity from protein lysates Support Protocol 1: Immunoprecipitation of Cdk5 using Dynabeads Alternate Protocol: Non-radioactive protocols to measure Cdk5 kinase activity Support Protocol 2: Western blot analysis for the detection of Cdk5, p35, and p39 Support Protocol 3: Immunodetection analysis for Cdk5, p35, and p39 Support Protocol 4: Genetically engineered mice (+ and - controls) Basic Protocol 2: Identifying new Cdk5 substrates and kinase inhibitors.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , Neuronas , Animales , Orientación del Axón , Quinasa 5 Dependiente de la Ciclina/metabolismo , Ratones , Neurogénesis , Neuronas/metabolismo , Fosforilación , Transducción de Señal
3.
Neuropsychopharmacology ; 44(6): 1177, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30787427

RESUMEN

The original version of this Article contained an error in the spelling of the author Anna K Radke, which was incorrectly given as Anna R Radke. This has now been corrected in both the PDF and HTML versions of the Article.

4.
Neuropsychopharmacology ; 44(6): 1163-1173, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30622300

RESUMEN

Obsessive-compulsive disorder (OCD) is a severe, chronic neuropsychiatric disorder with a strong genetic component. The SLC1A1 gene encoding the neuronal glutamate transporter EAAT3 has been proposed as a candidate gene for this disorder. Gene variants affecting SLC1A1 expression in human brain tissue have been associated with OCD. Several mouse models fully or partially lacking EAAT3 have shown no alterations in baseline anxiety-like or repetitive behaviors. We generated a transgenic mouse model (EAAT3glo) to achieve conditional, Cre-dependent EAAT3 overexpression and evaluated the overall impact of increased EAAT3 expression at behavioral and synaptic levels. Mice with EAAT3 overexpression driven by CaMKIIα-promoter (EAAT3glo/CMKII) displayed increased anxiety-like and repetitive behaviors that were both restored by chronic, but not acute, treatment with fluoxetine or clomipramine. EAAT3glo/CMKII mice also displayed greater spontaneous recovery of conditioned fear. Electrophysiological and biochemical analyses at corticostriatal synapses of EAAT3glo/CMKII mice revealed changes in NMDA receptor subunit composition and altered NMDA-dependent synaptic plasticity. By recapitulating relevant behavioral, neurophysiological, and psychopharmacological aspects, our results provide support for the glutamatergic hypothesis of OCD, particularly for the increased EAAT3 function, and provide a valuable animal model that may open novel therapeutic approaches to treat this devastating disorder.


Asunto(s)
Ansiedad/metabolismo , Conducta Animal/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Corteza Cerebral/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Neostriado/metabolismo , Plasticidad Neuronal/fisiología , Trastorno Obsesivo Compulsivo/metabolismo , Animales , Línea Celular , Clomipramina/farmacología , Modelos Animales de Enfermedad , Transportador 3 de Aminoácidos Excitadores/genética , Fluoxetina/farmacología , Expresión Génica/genética , Ratones , Ratones Transgénicos , Neuroblastoma , Técnicas de Placa-Clamp , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
5.
Front Physiol ; 9: 65, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29467671

RESUMEN

The participation of reactive oxygen species (ROS) generated by NOX1 and NOX2/NADPH oxidase has been documented during inflammatory pain. However, the molecular mechanism involved in their activation is not fully understood. We reported earlier a key role of Cyclin-dependent kinase 5 (Cdk5) during inflammatory pain. In particular, we demonstrated that TNF-α increased p35 expression, a Cdk5 activator, causing Cdk5-mediated TRPV1 phosphorylation followed by an increment in Ca2+ influx in nociceptive neurons and increased pain sensation. Here we evaluated if Cdk5 activation mediated by p35 transfection in HEK293 cells or by TNF-α treatment in primary culture of nociceptive neurons could increase ROS production. By immunofluorescence we detected the expression of catalytic subunit (Nox1 and Nox2) and their cytosolic regulators (NOXO1 and p47phox) of NOX1 and NOX2/NADPH oxidase complexes, and their co-localization with Cdk5/p35 in HEK293 cells and in nociceptive neurons. By using a hydrogen peroxide sensor, we detected a significant increase of ROS production in p35 transfected HEK293 cells as compared with control cells. This effect was significantly blocked by VAS2870 (NADPH oxidase inhibitor) or by roscovitine (Cdk5 activity inhibitor). Also by using another ROS probe named DCFH-DA, we found a significant increase of ROS production in nociceptive neurons treated with TNF-α and this effect was also blocked by VAS2870 or by roscovitine treatment. Interestingly, TNF-α increased immunodetection of p35 protein and NOX1 and NOX2/NADPH oxidase complexes in primary culture of trigeminal ganglia neurons. Finally, the cytosolic regulator NOXO1 was significantly translocated to plasma membrane after TNF-α treatment and roscovitine blocked this effect. Altogether these results suggest that Cdk5 activation is implicated in the ROS production by NOX1 and NOX2/NADPH oxidase complexes during inflammatory pain.

6.
Pain ; 158(11): 2155-2168, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28809765

RESUMEN

The purinergic P2X2 receptor (P2X2R) is an adenosine triphosphate-gated ion channel widely expressed in the nervous system. Here, we identified a putative cyclin-dependent kinase 5 (Cdk5) phosphorylation site in the full-size variant P2X2aR (TPKH), which is absent in the splice variant P2X2bR. We therefore investigated the effects of Cdk5 and its neuronal activator, p35, on P2X2aR function. We found an interaction between P2X2aR and Cdk5/p35 by co-immunofluorescence and co-immunoprecipitation in HEK293 cells. We also found that threonine phosphorylation was significantly increased in HEK293 cells co-expressing P2X2aR and p35 as compared to cells expressing only P2X2aR. Moreover, P2X2aR-derived peptides encompassing the Cdk5 consensus motif were phosphorylated by Cdk5/p35. Whole-cell patch-clamp recordings indicated a delay in development of use-dependent desensitization (UDD) of P2X2aR but not of P2X2bR in HEK293 cells co-expressing P2X2aR and p35. In Xenopus oocytes, P2X2aRs showed a slower UDD than in HEK293 cells and Cdk5 activation prevented this effect. A similar effect was found in P2X2a/3R heteromeric currents in HEK293 cells. The P2X2aR-T372A mutant was resistant to UDD. In endogenous cells, we observed similar distribution between P2X2R and Cdk5/p35 by co-localization using immunofluorescence in primary culture of nociceptive neurons. Moreover, co-immunoprecipitation experiments showed an interaction between Cdk5 and P2X2R in mouse trigeminal ganglia. Finally, endogenous P2X2aR-mediated currents in PC12 cells and P2X2/3R mediated increases of intracellular Ca in trigeminal neurons were Cdk5 dependent, since inhibition with roscovitine accelerated the desensitization kinetics of these responses. These results indicate that the P2X2aR is a novel target for Cdk5-mediated phosphorylation, which might play important physiological roles including pain signaling.


Asunto(s)
Activación del Canal Iónico/fisiología , Receptores Purinérgicos P2X2/metabolismo , Células Receptoras Sensoriales/fisiología , Treonina/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Ganglios Espinales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Ratones , Mutación/genética , Oocitos , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Ratas , Receptores Purinérgicos P2X2/genética , Receptores Purinérgicos P2X3/genética , Receptores Purinérgicos P2X3/metabolismo , Roscovitina , Células Receptoras Sensoriales/efectos de los fármacos , Treonina/genética , Xenopus
7.
Pain ; 157(6): 1346-1362, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26894912

RESUMEN

We reported earlier that TNF-α, a proinflammatory cytokine implicated in many inflammatory disorders causing orofacial pain, increases the activity of Cdk5, a key kinase involved in brain development and function and recently found to be involved in pain signaling. To investigate a potential mechanism underlying inflammatory pain in trigeminal ganglia (TGs), we engineered a transgenic mouse model (TNF) that can conditionally overexpresses TNF-α upon genomic recombination by Cre recombinase. TNF mice were bred with Nav1.8-Cre mouse line that expresses the Cre recombinase in sensory neurons to obtain TNF-α:Nav1.8-Cre (TNF-α cTg) mice. Although TNF-α cTg mice appeared normal without any gross phenotype, they displayed a significant increase in TNF-α levels after activation of NFκB signaling in the TG. IL-6 and MCP-1 levels were also increased along with intense immunostaining for Iba1 and GFAP in TG, indicating the presence of infiltrating macrophages and the activation of satellite glial cells. TNF-α cTg mice displayed increased trigeminal Cdk5 activity, and this increase was associated with elevated levels of phospho-T407-TRPV1 and capsaicin-evocated Ca influx in cultured trigeminal neurons. Remarkably, this effect was prevented by roscovitine, an inhibitor of Cdk5, which suggests that TNF-α overexpression induced sensitization of the TRPV1 channel. Furthermore, TNF-α cTg mice displayed more aversive behavior to noxious thermal stimulation (45°C) of the face in an operant pain assessment device as compared with control mice. In summary, TNF-α overexpression in the sensory neurons of TNF-α cTg mice results in inflammatory sensitization and increased Cdk5 activity; therefore, this mouse model would be valuable for investigating the mechanism of TNF-α involved in orofacial pain.


Asunto(s)
Calcio/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/metabolismo , Ganglio del Trigémino/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Quimiocina CCL2/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/genética
8.
PLoS One ; 10(2): e0117680, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25679528

RESUMEN

Prenatal stress causes predisposition to cognitive and emotional disturbances and is a risk factor towards the development of neuropsychiatric conditions like depression, bipolar disorders and schizophrenia. The extracellular protein Reelin, expressed by Cajal-Retzius cells during cortical development, plays critical roles on cortical lamination and synaptic maturation, and its deregulation has been associated with maladaptive conditions. In the present study, we address the effect of prenatal restraint stress (PNS) upon Reelin expression and signaling in pregnant rats during the last 10 days of pregnancy. Animals from one group, including control and PNS exposed fetuses, were sacrificed and analyzed using immunohistochemical, biochemical, cell biology and molecular biology approaches. We scored changes in the expression of Reelin, its signaling pathway and in the methylation of its promoter. A second group included control and PNS exposed animals maintained until young adulthood for behavioral studies. Using the optical dissector, we show decreased numbers of Reelin-positive neurons in cortical layer I of PNS exposed animals. In addition, neurons from PNS exposed animals display decreased Reelin expression that is paralleled by changes in components of the Reelin-signaling cascade, both in vivo and in vitro. Furthermore, PNS induced changes in the DNA methylation levels of the Reelin promoter in culture and in histological samples. PNS adult rats display excessive spontaneous locomotor activity, high anxiety levels and problems of learning and memory consolidation. No significant visuo-spatial memory impairment was detected on the Morris water maze. These results highlight the effects of prenatal stress on the Cajal-Retzius neuronal population, and the persistence of behavioral consequences using this treatment in adults, thereby supporting a relevant role of PNS in the genesis of neuropsychiatric diseases. We also propose an in vitro model that can yield new insights on the molecular mechanisms behind the effects of prenatal stress.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Metilación de ADN , Proteínas de la Matriz Extracelular/genética , Exposición Materna , Trastornos Mentales/etiología , Proteínas del Tejido Nervioso/genética , Efectos Tardíos de la Exposición Prenatal , Regiones Promotoras Genéticas , Serina Endopeptidasas/genética , Estrés Fisiológico , Estrés Psicológico , Animales , Animales Recién Nacidos , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Conducta Animal , Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Embarazo , Ratas , Proteína Reelina , Serina Endopeptidasas/metabolismo , Transducción de Señal
9.
PPAR Res ; 2014: 768594, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25246934

RESUMEN

Neuronal processes (neurites and axons) have an important role in brain cells communication and, generally, they are damaged in neurodegenerative diseases. Recent evidence has showed that the activation of PPARγ pathway promoted neuronal differentiation and axon polarity. In addition, activation of PPARγ using thiazolidinediones (TZDs) prevented neurodegeneration by reducing neuronal death, improving mitochondrial function, and decreasing neuroinflammation in neuropathic pain. In this review, we will discuss important evidence that supports a possible role of PPARγ in neuronal development, improvement of neuronal health, and pain signaling. Therefore, activation of PPARγ is a potential target with therapeutic applications against neurodegenerative disorders, brain injury, and pain regulation.

10.
PLoS One ; 9(3): e89310, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24662752

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Factores de Transcripción SOXD/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/citología , Encéfalo/embriología , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/deficiencia , Quinasa 5 Dependiente de la Ciclina/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Ratones , Mitosis/efectos de los fármacos , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosfotransferasas/metabolismo , Purinas/farmacología , Ratas , Roscovitina , Factores de Transcripción SOXD/química , Factores de Transcripción SOXD/genética
11.
PLoS One ; 9(3): e90363, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24658276

RESUMEN

Protein phosphorylation is the most common post-translational modification that regulates several pivotal functions in cells. Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase which is mostly active in the nervous system. It regulates several biological processes such as neuronal migration, cytoskeletal dynamics, axonal guidance and synaptic plasticity among others. In search for novel substrates of Cdk5 in the brain we performed quantitative phosphoproteomics analysis, isolating phosphoproteins from whole brain derived from E18.5 Cdk5+/+ and Cdk5-/- embryos, using an Immobilized Metal-Ion Affinity Chromatography (IMAC), which specifically binds to phosphorylated proteins. The isolated phosphoproteins were eluted and isotopically labeled for relative and absolute quantitation (iTRAQ) and mass spectrometry identification. We found 40 proteins that showed decreased phosphorylation at Cdk5-/- brains. In addition, out of these 40 hypophosphorylated proteins we characterized two proteins, :MARCKS (Myristoylated Alanine-Rich protein Kinase C substrate) and Grin1 (G protein regulated inducer of neurite outgrowth 1). MARCKS is known to be phosphorylated by Cdk5 in chick neural cells while Grin1 has not been reported to be phosphorylated by Cdk5. When these proteins were overexpressed in N2A neuroblastoma cell line along with p35, serine phosphorylation in their Cdk5 motifs was found to be increased. In contrast, treatments with roscovitine, the Cdk5 inhibitor, resulted in an opposite effect on serine phosphorylation in N2A cells and primary hippocampal neurons transfected with MARCKS. In summary, the results presented here identify Grin 1 as novel Cdk5 substrate and confirm previously identified MARCKS as a a bona fide Cdk5 substrate.


Asunto(s)
Encéfalo/metabolismo , Quinasa 5 Dependiente de la Ciclina/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Línea Celular , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Eliminación de Gen , Espectrometría de Masas , Ratones , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Fosfoproteínas/química , Fosforilación , Proteómica , Purinas/farmacología , Roscovitina
12.
J Neuroinflammation ; 11: 28, 2014 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-24495352

RESUMEN

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.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/deficiencia , Encefalitis/tratamiento farmacológico , Encefalitis/mortalidad , Encefalitis/patología , PPAR gamma/agonistas , Prosencéfalo/efectos de los fármacos , Tiazolidinedionas , Animales , Apoptosis/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Gliosis/etiología , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/patología , PPAR gamma/metabolismo , Fosfotransferasas/genética , Fosfotransferasas/metabolismo , Pioglitazona , Prosencéfalo/metabolismo , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico
13.
Mol Pain ; 9: 66, 2013 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-24359609

RESUMEN

BACKGROUND: Cyclin-dependent kinase 5 (Cdk5) is a unique member of the serine/threonine kinase family. This kinase plays an important role in neuronal development, and deregulation of its activity leads to neurodegenerative disorders. Cdk5 also serves an important function in the regulation of nociceptive signaling. Our previous studies revealed that the expression of Cdk5 and its activator, p35, is upregulated in nociceptive neurons during peripheral inflammation. The aim of the present study was to characterize the involvement of Cdk5 in orofacial pain. Since mechanical hyperalgesia is the distinctive sign of many orofacial pain conditions, we adapted an existing orofacial stimulation test to assess the behavioral responses to mechanical stimulation in the trigeminal region of the transgenic mice with either reduced or increased Cdk5 activity. RESULTS: Mice overexpressing or lacking p35, an activator of Cdk5, showed altered phenotype in response to noxious mechanical stimulation in the trigeminal area. Mice with increased Cdk5 activity displayed aversive behavior to mechanical stimulation as indicated by a significant decrease in reward licking events and licking time. The number of reward licking/facial contact events was significantly decreased in these mice as the mechanical intensity increased. By contrast, mice deficient in Cdk5 activity displayed mechanical hypoalgesia. CONCLUSIONS: Collectively, our findings demonstrate for the first time the important role of Cdk5 in orofacial mechanical nociception. Modulation of Cdk5 activity in primary sensory neurons makes it an attractive potential target for the development of novel analgesics that could be used to treat multiple orofacial pain conditions.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Hiperalgesia/enzimología , Animales , Quinasa 5 Dependiente de la Ciclina/genética , Dolor Facial/enzimología , Dolor Facial/metabolismo , Hiperalgesia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Ganglio del Trigémino/enzimología , Ganglio del Trigémino/metabolismo
14.
Mol Pain ; 9: 24, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23668392

RESUMEN

BACKGROUND: Odontoblasts are specialized cells that form dentin and they are believed to be sensors for tooth pain. Transforming growth factor-ß1 (TGF-ß1), a pro-inflammatory cytokine expressed early in odontoblasts, plays an important role in the immune response during tooth inflammation and infection. TGF-ß1 is also known to participate in pain signaling by regulating cyclin-dependent kinase 5 (Cdk5) in nociceptive neurons of the trigeminal and dorsal root ganglia. However, the precise role of TGF-ß1 in tooth pain signaling is not well characterized. The aim of our present study was to determine whether or not in odontoblasts Cdk5 is functionally active, if it is regulated by TGF-ß1, and if it affects the downstream pain receptor, transient receptor potential vanilloid-1 (TRPV1). RESULTS: We first determined that Cdk5 and p35 are indeed expressed in an odontoblast-enriched primary preparation from murine teeth. For the subsequent analysis, we used an odontoblast-like cell line (MDPC-23) and found that Cdk5 is functionally active in these cells and its kinase activity is upregulated during cell differentiation. We found that TGF-ß1 treatment potentiated Cdk5 kinase activity in undifferentiated MDPC-23 cells. SB431542, a specific inhibitor of TGF-ß1 receptor 1 (Tgfbr1), when co-administered with TGF-ß1, blocked the induction of Cdk5 activity. TGF-ß1 treatment also activated the ERK1/2 signaling pathway, causing an increase in early growth response-1 (Egr-1), a transcription factor that induces p35 expression. In MDPC-23 cells transfected with TRPV1, Cdk5-mediated phosphorylation of TRPV1 at threonine-407 was significantly increased after TGF-ß1 treatment. In contrast, SB431542 co-treatment blocked TRPV1 phosphorylation. Moreover, TGF-ß1 treatment enhanced both proton- and capsaicin-induced Ca²âº influx in TRPV1-expressing MDPC-23 cells, while co-treatment with either SB431542 or roscovitine blocked this effect. CONCLUSIONS: Cdk5 and p35 are expressed in a murine odontoblast-enriched primary preparation of cells from teeth. Cdk5 is also functionally active in odontoblast-like MDPC-23 cells. TGF-ß1 sensitizes TRPV1 through Cdk5 signaling in MDPC-23 cells, suggesting the direct involvement of odontoblasts and Cdk5 in dental nociceptive pain transduction.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Odontoblastos/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular , Quinasa 5 Dependiente de la Ciclina/genética , Ratones , Nociceptores/metabolismo , Dolor/metabolismo , Fosforilación , Fosfotransferasas/metabolismo , Canales Catiónicos TRPV/genética
15.
Neurochem Int ; 62(6): 848-53, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23416045

RESUMEN

Rap1 signaling is important for migration, differentiation, axonal growth, and during neuronal polarity. Rap1 can be activated by external stimuli, which in turn regulates specific guanine nucleotide exchange factors such as C3G, among others. Cdk5 functions are also important to neuronal migration and differentiation. Since we found that pharmacological inhibition of Cdk5 by using roscovitine reduced Rap1 protein levels in COS-7 cells and also C3G contains three putative phosphorylation sites for Cdk5, we examined whether the Cdk5-dependent phosphorylation of C3G could affect Rap1 expression and activity. We co-transfected C3G and tet-OFF system for p35 over-expression, an activator of Cdk5 activity into COS-7 cells, and then we evaluated phosphorylation in serine residues in C3G by immunoprecipitation and Western blot. We found that p35 over-expression increased C3G-serine-phosphorylation while inhibition of p35 expression by tetracycline or inhibition of Cdk5 activity with roscovitine decreased it. Interestingly, we found that MG-132, a proteasome inhibitor, rescue Rap1 protein levels in the presence of roscovitine. Besides, C3G-serine-phosphorylation and Rap1 protein levels were reduced in brain from Cdk5(-/-) as compared with the Cdk5(+/+) brain. Finally, we found that p35 over-expression increased Rap1 activity while inhibition of p35 expression by tetracycline or roscovitine decreased Rap1 activity. These results suggest that Cdk5-mediated serine-phosphorylation of C3G may control Rap1 stability and activity, and this may potentially impact various neuronal functions such as migration, differentiation, and polarity.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/fisiología , Proteínas de Unión al GTP rap1/fisiología , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Inhibidores de Cisteína Proteinasa , Factor 2 Liberador de Guanina Nucleótido/fisiología , Inmunoprecipitación , Subunidad p35 de la Interleucina-12/biosíntesis , Leupeptinas/farmacología , Proteínas de la Fusión de la Membrana , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Fosforilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Tetraciclina/farmacología , Transfección
16.
J Biol Chem ; 287(20): 16917-29, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22451679

RESUMEN

In addition to many important roles for Cdk5 in brain development and synaptic function, we reported previously that Cdk5 regulates inflammatory pain signaling, partly through phosphorylation of transient receptor potential vanilloid 1 (TRPV1), an important Na(+)/Ca(2+) channel expressed in primary nociceptive afferent nerves. Because TGF-ß regulates inflammatory processes and its receptor is expressed in TRPV1-positive afferents, we studied the cross-talk between these two pathways in sensory neurons during experimental peripheral inflammation. We demonstrate that TGF-ß1 increases transcription and protein levels of the Cdk5 co-activator p35 through ERK1/2, resulting in an increase in Cdk5 activity in rat B104 neuroblastoma cells. Additionally, TGF-ß1 enhances the capsaicin-induced Ca(2+) influx in cultured primary neurons from dorsal root ganglia (DRG). Importantly, Cdk5 activity was reduced in the trigeminal ganglia and DRG of 14-day-old TGF-ß1 knock-out mice, resulting in reduced Cdk5-dependent phosphorylation of TRPV1. The decreased Cdk5 activity is associated with attenuated thermal hyperalgesia in TGF-ß1 receptor conditional knock-out mice, where TGF-ß signaling is significantly reduced in trigeminal ganglia and DRG. Collectively, our results indicate that active cross-talk between the TGF-ß and Cdk5 pathways contributes to inflammatory pain signaling.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Ganglios Espinales/metabolismo , Sistema de Señalización de MAP Quinasas , Células Receptoras Sensoriales/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Ganglio del Trigémino/metabolismo , Animales , Calcio/metabolismo , Línea Celular Tumoral , Quinasa 5 Dependiente de la Ciclina/genética , Ganglios Espinales/patología , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/patología , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Dolor/genética , Dolor/metabolismo , Dolor/patología , Fosforilación/genética , Ratas , Células Receptoras Sensoriales/patología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Factor de Crecimiento Transformador beta1/genética , Ganglio del Trigémino/patología
17.
Cell Cycle ; 11(8): 1603-10, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22456337

RESUMEN

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.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Factores de Transcripción E2F/metabolismo , Neuronas/metabolismo , Proteína de Retinoblastoma/metabolismo , Secuencia de Aminoácidos , Animales , Quinasa 5 Dependiente de la Ciclina/deficiencia , Quinasa 5 Dependiente de la Ciclina/genética , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosfopéptidos/análisis , Fosforilación
18.
Cell Signal ; 24(1): 44-52, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21924349

RESUMEN

Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase that is mostly active in the nervous system, where it regulates several processes such as neuronal migration, actin and microtubule dynamics, axonal guidance, and synaptic plasticity, among other processes. In addition to these known functions, in the past few years, novel roles for Cdk5 outside of the nervous system have been proposed. These include roles in gene transcription, vesicular transport, apoptosis, cell adhesion, and migration in many cell types and tissues such as pancreatic cells, muscle cells, neutrophils, and others. In this review, we will summarize the recently studied non-neuronal functions of Cdk5, with a thorough analysis of the biological consequences of these novel roles.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/fisiología , Regulación de la Expresión Génica , Animales , Apoptosis , Adhesión Celular , Movimiento Celular , Senescencia Celular , Quinasa 5 Dependiente de la Ciclina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Desarrollo de Músculos , Neoplasias/metabolismo , Neovascularización Fisiológica , Fosforilación , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , Vesículas Transportadoras/metabolismo
19.
Mol Pain ; 7: 49, 2011 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-21736731

RESUMEN

BACKGROUND: We have previously reported that cyclin-dependent kinase 5 (Cdk5) participates in the regulation of nociceptive signaling. Through activation of the ERK1/2 pathway, Tumor Necrosis Factor-α (TNF-α) induces expression of Egr-1. This results in the sustained and robust expression of p35, a coactivator of Cdk5, in PC12 cells, thereby increasing Cdk5 kinase activity. The aim of our present study was to test whether resveratrol, a polyphenolic compound with known analgesic activity, can regulate Cdk5/p35 activity. RESULTS: Here we used a cell-based assay in which a p35 promoter-luciferase construct was stably transfected in PC12 cells. Our studies demonstrate that resveratrol inhibits p35 promoter activity and also blocks the TNF-α mediated increase in Cdk5 activity in PC12 cells. Resveratrol also inhibits p35 expression and blocks the TNF-α mediated increase in Cdk5 activity in DRG neurons. In the presence of resveratrol, the MEK inhibitor decreased p35 promoter activity, whereas the inhibitors of p38 MAPK, JNK and NF-κB increased p35 promoter activity, indicating that these pathways regulate p35 expression differently. The TNF-α-mediated increase in Egr-1 expression was decreased by resveratrol treatment with a concomitant reduction in p35 expression and protein levels, resulting in reduced Cdk5 kinase activity. CONCLUSIONS: We demonstrate here that resveratrol regulates p35 promoter activity in PC12 cells and DRG neurons. Most importantly, resveratrol blocks the TNF-α-mediated increase in p35 promoter activity, thereby reducing p35 expression and subsequent Cdk5 kinase activity. This new molecular mechanism adds to the known analgesic effects of resveratrol and confirms the need for identifying new analgesics based on their ability to inhibit Cdk5 activity for effective treatment of pain.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Fosfotransferasas/metabolismo , Estilbenos/farmacología , Animales , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/enzimología , Regulación de la Expresión Génica/efectos de los fármacos , Luciferasas/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Células PC12 , Fosfotransferasas/genética , Regiones Promotoras Genéticas/genética , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Resveratrol , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
20.
J Neurosci Res ; 89(9): 1489-98, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21671254

RESUMEN

Human T-cell leukemia virus type I (HTLV-I)-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a neurodegenerative disease characterized by selective loss of axons and myelin in the corticospinal tracts. This central axonopathy may originate from the impairment of anterograde axoplasmic transport. Previous work showed tau hyperphosphorylation at T(181) in cerebrospinal fluid of HAM/TSP patients. Similar hyperphosphorylation occurs in SH-SY5Y cells incubated with supernatant from MT-2 cells (HTLV-I-infected lymphocytes secreting viral proteins, including Tax) that produce neurite shortening. Tau phosphorylation at T(181) is attributable to glycogen synthase kinase 3-ß (GSK3-ß) and cyclin-dependent kinase 5 (CDK5) activation. Here we investigate whether neurite retraction in the SH-SY5Y model associates with concurrent changes in other tau hyperphosphorylable residues. Threonine 181 turned out to be the only tau hyperphosphorylated residue. We also evaluate the role of GSK3-ß and CDK5 in this process by using specific kinase inhibitors (LiCl, TDZD-8, and roscovitine). Changes in both GSK3-ß active and inactive forms were followed by measuring the regulatory phosphorylable sites (S(9) and Y(216) , inactivating and activating phosphorylation, respectively) together with changes in ß-catenin protein levels. Our results showed that LiCl and TDZD-8 were unable to prevent MT-2 supernatant-mediated neurite retraction and also that neither Y(216) nor S(9) phosphorylations were changed in GSK3-ß. Thus, GSK3-ß seems not to play a role in T(181) hyperphosphorylation. On the other hand, the CDK5 involvement in tau phosphorylation was confirmed by both the increase in its enzymatic activity and the absence of MT-2 neurite retraction in the presence of roscovitine or CDK5 siRNA transfection.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Neuritas/efectos de los fármacos , Enfermedades Neurodegenerativas/virología , Linfocitos T/virología , Análisis de Varianza , Factores Biológicos/metabolismo , Factores Biológicos/fisiología , Medios de Cultivo Condicionados/farmacología , Productos del Gen tax/metabolismo , Productos del Gen tax/farmacología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Neuritas/enzimología , Neuritas/inmunología , Neuritas/patología , Neuroblastoma , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/patología , Fosforilación/efectos de los fármacos , Estadísticas no Paramétricas , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...