Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 636(Pt 1): 162-169, 2022 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-36334440

RESUMEN

Primary cilia transduce signals via transmembrane and membrane-associated proteins localized to the ciliary membrane in vertebrate cells. In humans, transmembrane protein 67 (TMEM67), a component of the multiprotein complex functioning as a gatekeeper at the transition zone (TZ) of primary cilia, is mutated in patients suffering from cilia-related pleiotropic diseases, collectively referred to as ciliopathies. The requirement of TMEM67 for the gating function of the TZ that delivers membrane proteins into the ciliary compartment has not been determined. In this study, we established hTERT-RPE1 cells with knockout (KO) of TMEM67 and examined whether cilium formation and TZ gating are affected by its ablation. TMEM67-KO cells displayed impaired ciliogenesis, elongated cilia, perturbed ciliary localization of membrane-associated proteins ARL13B and INPP5E but normal recruitment of TZ proteins CEP290, RPGRIP1L and NPHP5. The exogenous expression of ciliopathy-associated TMEM67 mutants restored ciliary localization of ARL13B and INPP5E but failed to attenuate aberrant cilium elongation in TMEM67-KO cells. Furthermore, we found that TMEM67 localization is not confined to the TZ but extends into the cilium. Our findings indicate that TMEM67 is required not only for ciliogenesis and cilium length regulation but also for the gating function of the TZ independently of RPGRIP1L/CEP290/NPHP5 recruitment to this region. They further suggest that aberrant cilium elongation underlies the pathogenesis of TMEM67-linked ciliopathies.


Asunto(s)
Cilios , Ciliopatías , Humanos , Cilios/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Ciliopatías/genética , Ciliopatías/metabolismo , Antígenos de Neoplasias/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Ciclo Celular/metabolismo , Factores de Ribosilacion-ADP/metabolismo
2.
Neurochem Int ; 153: 105273, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34971749

RESUMEN

The axon initial segment (AIS) is a structural neuronal compartment of the proximal axon that plays key roles in sodium channel clustering, action potential initiation, and signal propagation of neuronal outputs. Mutations in constitutive genes of the AIS, such as ANK3, have been identified in patients with neurodevelopmental disorders. Nevertheless, morphological changes in the AIS in neurodevelopmental disorders have not been characterized. In this study, we investigated the length of the AIS in rodent models of attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). We observed abnormalities in AIS length in both animal models. In ADHD model rodents, we observed shorter AIS length in layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC) and primary somatosensory barrel field (S1BF). Further, we observed shorter AIS length in S1BF L5 neurons. In ASD model mice, we observed shorter AIS length in L2/3 and L5 neurons of the S1BF. These results suggest that impairments in AIS length are common phenomena in neurodevelopmental disorders such as ADHD and ASD and may be conserved across species. Our findings provide novel insight into the potential contribution of the AIS to the pathophysiology and pathogenesis of neurodevelopmental disorders.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Segmento Inicial del Axón , Trastornos del Neurodesarrollo , Animales , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno del Espectro Autista/genética , Humanos , Ratones , Roedores
3.
Mol Biol Cell ; 31(18): 1963-1973, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32583741

RESUMEN

Arginine methylation is a common posttranslational modification that modulates protein function. SCY1-like pseudokinase 1 (SCYL1) is crucial for neuronal functions and interacts with γ2-COP to form coat protein complex I (COPI) vesicles that regulate Golgi morphology. However, the molecular mechanism by which SCYL1 is regulated remains unclear. Here, we report that the γ2-COP-binding site of SCYL1 is arginine-methylated by protein arginine methyltransferase 1 (PRMT1) and that SCYL1 arginine methylation is important for the interaction of SCYL1 with γ2-COP. PRMT1 was colocalized with SCYL1 in the Golgi fraction. Inhibition of PRMT1 suppressed axon outgrowth and dendrite complexity via abnormal Golgi morphology. Knockdown of SCYL1 by small interfering RNA (siRNA) inhibited axon outgrowth, and the inhibitory effect was rescued by siRNA-resistant SCYL1, but not SCYL1 mutant, in which the arginine methylation site was replaced. Thus, PRMT1 regulates Golgi morphogenesis via SCYL1 arginine methylation. We propose that SCYL1 arginine methylation by PRMT1 contributes to axon and dendrite morphogenesis in neurons.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteína Coatómero/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Animales , Arginina/metabolismo , Proteína Coat de Complejo I/metabolismo , Proteína Coatómero/fisiología , Proteínas de Unión al ADN/fisiología , Femenino , Aparato de Golgi/metabolismo , Células HEK293 , Células HeLa , Humanos , Masculino , Metilación , Ratones , Ratones Endogámicos ICR , Proyección Neuronal/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/fisiología , Ratas , Ratas Wistar , Proteínas Represoras/fisiología , Factores de Transcripción/metabolismo
4.
Biochem Biophys Res Commun ; 509(1): 227-234, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30583862

RESUMEN

The primary cilia are known as biosensors that transduce signals through the ciliary membrane proteins in vertebrate cells. The ciliary membrane contains transmembrane proteins and membrane-associated proteins. Tubby-like protein 3 (TULP3), a member of the tubby family, has been shown to interact with the intraflagellar transport-A complex (IFT-A) and to be involved in the ciliary localization of transmembrane proteins, although its role in the ciliary entry of membrane-associated proteins has remained unclear. Here, to determine whether TULP3 is required for the localization of ciliary membrane-associated proteins, we generated and analyzed TULP3-knockout (KO) hTERT RPE-1 (RPE1) cells. Immunofluorescence analysis demonstrated that ciliary formation was downregulated in TULP3-KO cells and that membrane-associated proteins, ADP-ribosylation factor-like 13B (ARL13B) and inositol polyphosphate-5-phosphatase E (INPP5E), failed to localize to primary cilia in TULP3-KO cells. These defects in the localization of ARL13B and INPP5E in TULP3-KO cells were rescued by the exogenous expression of wild-type TULP3, but not that of mutant TULP3 lacking the ability to bind IFT-A. In addition, the expression of TUB protein, another member of the tubby family whose endogenous expression is absent in RPE1 cells, also rescued the defective ciliary localization of ARL13B and INPP5E in TULP3-KO cells, suggesting that there is functional redundancy between TULP3 and TUB. Our findings indicate that TULP3 participates in ciliogenesis, and targets membrane-associated proteins to primary cilia via binding to IFT-A.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Cilios/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas/metabolismo , Factores de Ribosilacion-ADP/análisis , Sistemas CRISPR-Cas , Proteínas Portadoras/análisis , Proteínas Portadoras/metabolismo , Línea Celular , Cilios/genética , Cilios/ultraestructura , Técnicas de Inactivación de Genes , Humanos , Péptidos y Proteínas de Señalización Intracelular , Monoéster Fosfórico Hidrolasas/análisis , Unión Proteica , Proteínas/genética
5.
PLoS One ; 10(4): e0123578, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25928708

RESUMEN

Recently, accumulating reports have suggested the importance of endoplasmic reticulum (ER) stress signaling in the differentiation of several tissues and cells, including myoblasts and osteoblasts. Secretory cells are easily subjected to ER stress during maturation of their secreted proteins. Skin fibroblasts produce and release several proteins, such as collagens, matrix metalloproteinases (MMPs), the tissue inhibitors of metalloproteinases (TIMPs) and glycosaminoglycans (GAGs), and the production of these proteins is increased at wound sites. Differentiation of fibroblasts into myofibroblasts is one of the key factors for wound healing and that TGF-ß can induce fibroblast differentiation into myofibroblasts, which express α-smooth muscle actin. Well-differentiated myofibroblasts show increased production of collagen and TGF-ß, and bring about wound healing. In this study, we examined the effects of ER stress signaling on the differentiation of fibroblasts, which is required for wound healing, using constitutively ER stress-activated primary cultured fibroblasts. The cells expressed positive α-smooth muscle actin signals without TGF-ß stimulation compared with control fibroblasts. Gel-contraction assays suggested that ER stress-treated primary fibroblasts caused stronger shrinkage of collagen gels than control cells. These results suggest that ER stress signaling could accelerate the differentiation of fibroblasts to myofibroblasts at injured sites. The present findings may provide important insights for developing therapies to improve wound healing.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Miofibroblastos/citología , Miofibroblastos/metabolismo , Animales , Western Blotting , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Colágeno/metabolismo , Estrés del Retículo Endoplásmico/genética , Fibroblastos , Glicosaminoglicanos/metabolismo , Inmunohistoquímica , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta/metabolismo
6.
Neurochem Res ; 40(6): 1165-78, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25894684

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms that precede the onset of motor symptoms. Rotenone is often used to induce PD-like pathology in the central nervous system (CNS) and enteric nervous system (ENS). However, there is little or no information on the temporal changes in other neural tissues and the spread of pathology throughout the entire body organs. Here, we recorded the serial immunohistochemical changes in neurons and glial cells of the striatum, substantia nigra (SN), olfactory bulb (OB), thoracic cord (ThC) and ascending colon (AC) induced by 1-, 3- and 6-week administration of rotenone (50 mg/kg/day) infused subcutaneously in C57BL mice using an osmotic pump. Rotenone exposure for 3 or 6 weeks caused neurodegeneration in the striatum, whereas neuronal damage was seen in the SN and OB only after 6 weeks. Moreover, rotenone induced neurodegeneration in the myenteric plexus of AC but not in ThC. Rotenone also activated glial cells before any apparent neurodegeneration in the CNS but not in the ENS. Our results demonstrated that subcutaneous administration of rotenone can cause progressive neurodegeneration in the OB and AC, in addition to the nigrostriatal pathway, and temporal differential glial activation, and that these changes do not spread retrogradely from OB or ENS to nigrostriatal pathway. The results suggested that the different vulnerability of neurons to the neurotoxic effects of rotenone administrated subcutaneously are due to glial activation in these neural tissues.


Asunto(s)
Sistema Nervioso Central/patología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Sistema Nervioso Periférico/patología , Rotenona/toxicidad , Desacopladores/toxicidad , Animales , Encéfalo/patología , Colon Ascendente/patología , Neuronas Dopaminérgicas/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/patología , Vías Nerviosas/patología , Neuronas/patología
7.
Acta Med Okayama ; 68(6): 317-22, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25519025

RESUMEN

In vertebrates, almost all somatic cells extend a single immotile cilium, referred to as a primary cilium. Increasing evidence suggests that primary cilia serve as cellular antennae in many types of tissues by sensing chemical or mechanical stimuli in the milieu surrounding the cells. In rodents an antibody to adenylyl cyclase 3 (AC3) has been widely used to label the primary cilia of neurons in vivo by immunostaining, whereas the lack of markers for the primary cilia of astrocytes has made it difficult to observe astrocytic primary cilia in vivo. Here, we obtained a visualization of astrocytic primary cilia in the mouse brain. In the somatosensory cortex, a large portion of neurons and astrocytes at postnatal day 10 (P10), and of neurons at P56 had AC3-positive primary cilia, whereas only approx. one-half of the astrocytes in the P56 mice carried primary cilia weakly positive for AC3. In contrast, the majority of astrocytes had ADP-ribosylation factor-like protein 13B (Arl13b)-positive primary cilia in the somatosensory cortex and other brain regions of P56 mice. The lengths of astrocytic primary cilia positive for Arl13b varied among the brain regions. Our data indicate that Arl13b is a noteworthy marker of astrocytic primary cilia in the brain.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Cilios/metabolismo , Animales , Astrocitos/citología , Biomarcadores/metabolismo , Encéfalo/citología , Cilios/ultraestructura , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Modelos Animales , Neuronas/citología , Neuronas/metabolismo , Corteza Somatosensorial/citología , Corteza Somatosensorial/metabolismo
8.
Neurotox Res ; 26(3): 285-98, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24923464

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease with motor symptoms as well as non-motor symptoms that precede the onset of motor symptoms. Mitochondrial complex I inhibitor, rotenone, has been widely used to reproduce PD pathology in the central nervous system (CNS) and enteric nervous system (ENS). We reported previously that metallothioneins (MTs) released from astrocytes can protect dopaminergic neurons against oxidative stress. The present study examined the changes in MT expression by chronic systemic rotenone administration in the striatum and colonic myenteric plexus of C57BL mice. In addition, we investigated the effects of MT depletion on rotenone-induced neurodegeneration in CNS and ENS using MT-1 and MT-2 knockout (MT KO) mice, or using primary cultured neurons from MT KO mice. In normal C57BL mice, subcutaneous administration of rotenone for 6 weeks caused neurodegeneration, increased MT expression with astrocytes activation in the striatum and myenteric plexus. MT KO mice showed more severe myenteric neuronal damage by rotenone administration after 4 weeks than wild-type mice, accompanied by reduced astroglial activation. In primary cultured mesencephalic neurons from MT KO mice, rotenone exposure induced neurotoxicity in dopaminergic neurons, which was complemented by addition of recombinant protein. The present results suggest that MT seems to provide protection against neurodegeneration in ENS of rotenone-induced PD model mice.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Metalotioneína/metabolismo , Plexo Mientérico/efectos de los fármacos , Fármacos Neuroprotectores/metabolismo , Trastornos Parkinsonianos/metabolismo , Rotenona/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Células Cultivadas , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Masculino , Metalotioneína/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Plexo Mientérico/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Trastornos Parkinsonianos/inducido químicamente
9.
PLoS One ; 9(5): e97918, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24830745

RESUMEN

In the rodent brain, certain G protein-coupled receptors and adenylyl cyclase type 3 are known to localize to the neuronal primary cilium, a primitive sensory organelle protruding singly from almost all neurons. A recent chemical screening study demonstrated that many compounds targeting dopamine receptors regulate the assembly of Chlamydomonas reinhardtii flagella, structures which are analogous to vertebrate cilia. Here we investigated the effects of dopaminergic inputs loss on the architecture of neuronal primary cilia in the rodent striatum, a brain region that receives major dopaminergic projections from the midbrain. We first analyzed the lengths of neuronal cilia in the dorsolateral striatum of hemi-parkinsonian rats with unilateral lesions of the nigrostriatal dopamine pathway. In these rats, the striatal neuronal cilia were significantly longer on the lesioned side than on the non-lesioned side. In mice, the repeated injection of reserpine, a dopamine-depleting agent, elongated neuronal cilia in the striatum. The combined administration of agonists for dopamine receptor type 2 (D2) with reserpine attenuated the elongation of striatal neuronal cilia. Repeated treatment with an antagonist of D2, but not of dopamine receptor type 1 (D1), elongated the striatal neuronal cilia. In addition, D2-null mice displayed longer neuronal cilia in the striatum compared to wild-type controls. Reserpine treatment elongated the striatal neuronal cilia in D1-null mice but not in D2-null mice. Repeated treatment with a D2 agonist suppressed the elongation of striatal neuronal cilia on the lesioned side of hemi-parkinsonian rats. These results suggest that the elongation of striatal neuronal cilia following the lack of dopaminergic inputs is attributable to the absence of dopaminergic transmission via D2 receptors. Our results provide the first evidence that the length of neuronal cilia can be modified by the lack of a neurotransmitter's input.


Asunto(s)
Cilios/patología , Neuronas Dopaminérgicas/patología , Enfermedad de Parkinson Secundaria/patología , Estriado Ventral/patología , Animales , Astrocitos/patología , Forma de la Célula , Agonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2/farmacología , Neuronas Dopaminérgicas/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Enfermedad de Parkinson Secundaria/metabolismo , Ratas Sprague-Dawley , Reserpina/farmacología , Sustancia Negra/patología
10.
Neurobiol Dis ; 59: 244-56, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23959140

RESUMEN

Astrocytes are abundant neuron-supporting glial cells that harbor a powerful arsenal of neuroprotective antioxidative molecules and neurotrophic factors. Here we examined whether enrichment with healthy striatal astrocytes can provide neuroprotection against progressive dopaminergic neurodegeneration. Serotonin 1A (5-HT1A) agonist 8-OH-DPAT induced astrocyte proliferation and increased metallothionein-1/-2 (MT-1/-2), antioxidative molecules, in cultured astrocytes and the striatum of mice. Primary cultured mesencephalic dopamine neurons were protected against oxidative stress by preincubation with conditioned media from 8-OH-DPAT-treated astrocytes. These protective effects were canceled by 5-HT1A antagonist or MT-1/-2-specific antibody. Furthermore, reduction of nigrostriatal dopaminergic neurons in 6-hydroxydopamine-lesioned parkinsonian model mice was significantly abrogated by repeated injections of 8-OH-DPAT. Treatment with 8-OH-DPAT markedly increased the expression of MT in striatal astrocytes in the hemi-parkinsonian mice. Our study provides a promising therapeutic strategy of neuroprotection against oxidative stress and progressive dopaminergic neurodegeneration by demonstrating the efficacy of targeting 5-HT1A receptors in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Neuronas Dopaminérgicas/fisiología , Enfermedad de Parkinson/patología , Receptor de Serotonina 5-HT1A/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralin/administración & dosificación , Adrenérgicos/toxicidad , Animales , Astrocitos/efectos de los fármacos , Encéfalo/citología , Buspirona/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Femenino , Lateralidad Funcional/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/prevención & control , Embarazo , Ratas , Ratas Sprague-Dawley , Agonistas de Receptores de Serotonina/administración & dosificación , Agua/farmacología
11.
Neurochem Res ; 37(9): 1944-51, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22674083

RESUMEN

Prostaglandin H synthase exerts not only cyclooxygenase activity but also peroxidase activity. The latter activity of the enzyme is thought to couple with oxidation of dopamine to dopamine quinone. Therefore, it has been proposed that cyclooxygenase inhibitors could suppress dopamine quinone formation. In the present study, we examined effects of various cyclooxygenase inhibitors against excess methyl L-3,4-dihydroxyphenylalanine (L-DOPA)-induced quinoprotein (protein-bound quinone) formation and neurotoxicity using dopaminergic CATH.a cells. The treatment with aspirin inhibited excess methyl L-DOPA-induced quinoprotein formation and cell death. However, acetaminophen did not show protective effects, and indomethacin and meloxicam rather aggravated these methyl L-DOPA-induced changes. Aspirin and indomethacin did not affect the level of glutathione that exerts quenching dopamine quinone in dopaminergic cells. In contrast with inhibiting effects of higher dose in the previous reports, relatively lower dose of aspirin that affected methyl L-DOPA-induced quinoprotein formation and cell death failed to prevent cyclooxygenase-induced dopamine chrome generation in cell-free system. Furthermore, aspirin but not acetaminophen or meloxicam showed direct dopamine quinone-scavenging effects in dopamine-semiquinone generating systems. The present results suggest that cyclooxygenase shows little contribution to dopamine oxidation in dopaminergic cells and that protective effects of aspirin against methyl L-DOPA-induced dopamine quinone neurotoxicity are based on its cyclooxygenase-independent property.


Asunto(s)
Aspirina/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Dopaminérgicos/toxicidad , Levodopa/antagonistas & inhibidores , Levodopa/toxicidad , Fármacos Neuroprotectores , Quinonas/toxicidad , Acetaminofén/farmacología , Analgésicos no Narcóticos/farmacología , Animales , Sistema Libre de Células , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Glutatión/metabolismo , Indometacina/farmacología , Meloxicam , Metildopa/toxicidad , Ratones , Neuronas/efectos de los fármacos , Quinonas/antagonistas & inhibidores , Simpaticolíticos/toxicidad , Tiazinas/farmacología , Tiazoles/farmacología
12.
Acta Med Okayama ; 65(5): 279-85, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22037264

RESUMEN

Almost all mammalian cells carry one primary cilium that functions as a biosensor for chemical and mechanical stimuli. Genetic damages that compromise cilia formation or function cause a spectrum of disorders referred to as ciliapathies. Recent studies have demonstrated that some pharmacological agents and extracellular environmental changes can alter primary cilium length. Renal injury is a well-known example of an environmental insult that triggers cilia length modification. Lithium treatment causes primary cilia to extend in several cell types including neuronal cells;this phenomenon is likely independent of glycogen synthase kinase-3ß inhibition. In renal epithelial cell lines, deflection of the primary cilia by fluid shear shortens them by reducing the intracellular cyclic AMP level, leading to a subsequent decrease in mechanosensitivity to fluid shear. Primary cilium length is also influenced by the dynamics of actin filaments and microtubules through the levels of soluble tubulin in the cytosol available for primary cilia extension. Thus, mammalian cells can adapt to the extracellular environment by modulating the primary cilium length, and this feedback system utilizing primary cilia might exist throughout the mammalian body. Further investigation is required concerning the precise molecular mechanisms underlying the control of primary cilium length in response to environmental factors.


Asunto(s)
Cilios/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Animales , Transporte Biológico , Línea Celular , Cilios/efectos de los fármacos , Cilios/metabolismo , AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Humanos , Riñón/citología , Riñón/lesiones , Litio/farmacología
13.
Glia ; 59(3): 435-51, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264950

RESUMEN

Our previous studies demonstrated the involvement of quinone formation in dopaminergic neuron dysfunction in the L-DOPA-treated parkinsonian model and in methamphetamine (METH) neurotoxicity. We further reported that the cysteine-rich metal-binding metallothionein (MT) family of proteins protects dopaminergic neurons against dopamine (DA) quinone neurotoxicity by its quinone-quenching property. The aim of this study was to examine MT induction in astrocytes in response to excess DA and the potential neuroprotective effects of astrocyte-derived MTs against DA quinone toxicity. DA exposure significantly upregulated MT-1/-2 in cultured striatal astrocytes, but not in mesencephalic neurons. This DA-induced MT upregulation in astrocytes was blocked by treatment with a DA-transporter (DAT) inhibitor, but not by DA-receptor antagonists. Expression of nuclear factor erythroid 2-related factor (Nrf2) and its binding activity to antioxidant response element of MT-1 gene were significantly increased in the astrocytes after DA exposure. Nuclear translocation of Nrf2 was suppressed by the DAT inhibitor. Quinone formation and reduction of mesencephalic DA neurons after DA exposure were ameliorated by preincubation with conditioned media from DA-treated astrocytes. These protective effects were abrogated by MT-1/-2-specific antibody. Adding exogenous MT-1 to glial conditioned media also showed similar neuroprotective effects. Furthermore, MT-1/-2 expression was markedly elevated specifically in reactive astrocytes in the striatum of L-DOPA-treated hemi-parkinsonian mice or METH-injected mice. These results suggested that excess DA taken up by astrocytes via DAT upregulates MT-1/-2 expression specifically in astrocytes, and that MTs or related molecules secreted specifically by astrocytes protect dopaminergic neurons from damage through quinone quenching and/or scavenging of free radicals.


Asunto(s)
Astrocitos/fisiología , Dopamina/análogos & derivados , Dopamina/fisiología , Metalotioneína/metabolismo , Metalotioneína/fisiología , Neuronas/metabolismo , Fármacos Neuroprotectores/toxicidad , Animales , Astrocitos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Dopamina/toxicidad , Depuradores de Radicales Libres/metabolismo , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley
14.
Ann Neurol ; 67(2): 239-49, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20225289

RESUMEN

OBJECTIVE: Recent double-blind, controlled trials in Japan showed that the antiepileptic agent zonisamide (ZNS) improves the cardinal symptoms of Parkinson's disease. Glutathione (GSH) exerts antioxidative activity through quenching reactive oxygen species and dopamine quinone. GSH depletion within dopaminergic neurons impairs mitochondrial complex I activity, followed by age-dependent nigrostriatal neurodegeneration. This study examined changes in GSH and GSH synthesis-related molecules, and the neuroprotective effects of ZNS on dopaminergic neurodegeneration using 6-hydroxydopamine-injected hemiparkinsonian mice brain and cultured neurons or astrocytes. METHODS AND RESULTS: ZNS increased both the cell number and GSH levels in astroglial C6 cells, but not in dopaminergic neuronal CATH.a cells. Repeated injections of ZNS (30mg/kg intraperitoneally) for 14 days also significantly increased GSH levels and S100beta-positive astrocytes in mouse basal ganglia. Repeated ZNS injections (30mg/kg) for 7 days in the hemiparkinsonian mice increased the expression of cystine/glutamate exchange transporter xCT in activated astrocytes, which supply cysteine to neurons for GSH synthesis. Treatment of these mice with ZNS also increased GSH levels and completely suppressed striatal levodopa-induced quinone formation. Reduction of nigrostriatal dopamine neurons in the lesioned side of hemiparkinsonian mice was significantly abrogated by repeated injections of ZNS with or without adjunctive levodopa starting 3 weeks after 6-hydroxydopamine lesioning. INTERPRETATION: These results provide new pharmacological evidence for the effects of ZNS. ZNS markedly increased GSH levels by enhancing the astroglial cystine transport system and/or astroglial proliferation via S100beta production or secretion. ZNS acts as a neuroprotectant against oxidative stress and progressive dopaminergic neurodegeneration.


Asunto(s)
Astrocitos/efectos de los fármacos , Isoxazoles/farmacología , Isoxazoles/uso terapéutico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Antiparkinsonianos/uso terapéutico , Astrocitos/metabolismo , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Carbidopa/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutatión/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Factores de Crecimiento Nervioso/metabolismo , Oxidopamina/efectos adversos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Zonisamida
15.
Biochem Biophys Res Commun ; 388(4): 757-62, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19703416

RESUMEN

The molecular mechanisms underlying the therapeutic effects of lithium, a first-line antimanic mood stabilizer, have not yet been fully elucidated. Treatment of the algae Chlamydomonas reinhardtii with lithium has been shown to induce elongation of their flagella, which are analogous structures to vertebrate cilia. In the mouse brain, adenylyl cyclase 3 (AC3) and certain neuropeptide receptors colocalize to the primary cilium of neuronal cells, suggesting a chemosensory function for the primary cilium in the nervous system. Here we show that lithium treatment elongates primary cilia in the mouse brain and in cultured cells. Brain sections from mice chronically fed with Li(2)CO(3) were subjected to immunofluorescence study. Primary cilia carrying both AC3 and the receptor for melanin-concentrating hormone (MCH) were elongated in the dorsal striatum and nucleus accumbens of lithium-fed mice, as compared to those of control animals. Moreover, lithium-treated NIH3T3 cells and cultured striatal neurons exhibited elongation of the primary cilia. The present results provide initial evidence that a psychotropic agent can affect ciliary length in the central nervous system, and furthermore suggest that lithium exerts its therapeutic effects via the upregulation of cilia-mediated MCH sensing. These findings thus contribute novel insights into the pathophysiology of bipolar mood disorder and other psychiatric diseases.


Asunto(s)
Antimaníacos/farmacología , Encéfalo/efectos de los fármacos , Carbonato de Litio/farmacología , Neuronas/efectos de los fármacos , Animales , Encéfalo/citología , Células Cultivadas , Cilios/efectos de los fármacos , Cilios/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Neuronas/citología
16.
FASEB J ; 23(10): 3289-97, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19470799

RESUMEN

The Drosophila pericentrin-like protein has been shown to be essential for the formation of the sensory cilia of chemosensory and mechanosensory neurons by mutant analysis in flies, while the in vivo function of pericentrin, a well-studied mammalian centrosomal protein related to microcephalic primordial dwarfism, has been unclear. To determine whether pericentrin is required for ciliogenesis in mammals, we generated and analyzed mice with a hypomorphic mutation of Pcnt encoding the mouse pericentrin. Immunofluorescence analysis demonstrated that olfactory cilia of chemosensory neurons in the nasal olfactory epithelium were malformed in the homozygous mutant mice. On the other hand, the assembly of motile and primary cilia of non-neuronal epithelial cells and the formation of sperm flagella were not affected in the Pcnt-mutant mice. The defective assembly of olfactory cilia in the mutant was apparent from birth. The mutant animals displayed reduced olfactory performance in agreement with the compromised assembly of olfactory cilia. Our findings suggest that pericentrin is essential for the assembly of chemosensory cilia of olfactory receptor neurons, but it is not globally required for cilia formation in mammals.


Asunto(s)
Antígenos/metabolismo , Centrosoma/metabolismo , Enanismo/genética , Microcefalia/genética , Bulbo Olfatorio/anomalías , Animales , Antígenos/genética , Cilios/metabolismo , Ratones , Ratones Mutantes , Transcripción Genética
17.
Am J Med Genet B Neuropsychiatr Genet ; 150B(7): 967-76, 2009 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-19191256

RESUMEN

Disrupted-in-Schizophrenia 1 (DISC1) and its molecular cascade have been implicated in the pathophysiology of major psychoses. Previously, we identified pericentrin 2 (PCNT2) and DISC1-binding zinc finger protein (DBZ) as binding partners of DISC1; further, we observed elevated expression of PCNT2 in the postmortem brains and in the lymphocytes of bipolar disorder patients, compared to controls. Here, we examined the association of PCNT2 with schizophrenia in a case-control study of Japanese cohorts. We also examined the association of DBZ with schizophrenia and with bipolar disorder, and compared the mRNA levels of DBZ in the postmortem brains of schizophrenia, bipolar and control samples. DNA from 180 schizophrenia patients 201 controls were used for the association study of PCNT2 and DBZ with schizophrenia. Association of DBZ with bipolar disorder was examined in DNA from 238 bipolar patients and 240 age- and gender-matched controls. We observed significant allelic and genotypic associations of the PCNT2 SNPs, rs2249057, rs2268524, and rs2073380 (Ser/Arg) with schizophrenia; the association of rs2249057 (P = 0.002) withstand multiple testing correction. Several two SNP- and three SNP-haplotypes showed significant associations; the associations of haplotypes involving rs2249057 withstand multiple testing correction. No associations were observed for DBZ with schizophrenia or with bipolar disorder; further, there was no significant difference between the DBZ mRNA levels of control, schizophrenia and bipolar postmortem brains. We suggest a possible role of PCNT2 in the pathogenesis of schizophrenia. Abnormalities of PCNT2, the centrosomal protein essential for microtubule organization, may be suggested to lead to neurodevelopmental abnormalities.


Asunto(s)
Antígenos/genética , Trastorno Bipolar/genética , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Proteínas del Tejido Nervioso/metabolismo , Esquizofrenia/genética , Factores de Transcripción/genética , Adulto , Alelos , Estudios de Casos y Controles , Demografía , Femenino , Genoma Humano/genética , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética , Unión Proteica
18.
Neurochem Res ; 34(4): 764-74, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18946735

RESUMEN

We examined changes in nuclear peroxisome proliferator-activated receptor gamma (PPAR gamma) in the striatum in methamphetamine (METH)-induced dopaminergic neurotoxicity, and also examined effects of treatment with drugs possessing PPAR gamma agonistic properties. The marked reduction of nuclear PPAR gamma-expressed cells was seen in the striatum 3 days after METH injections (4 mg/kg x 4, i.p. with 2-h interval). The reduction of dopamine transporter (DAT)-positive signals and PPAR gamma expression, and accumulation of activated microglial cells were significantly and dose-dependently attenuated by four injections of a nonsteroidal anti-inflammatory drug and a PPAR gamma ligand, ibuprofen (10 or 20 mg/kg x 4, s.c.) given 30 min prior to each METH injection, but not by either a low or high dose of aspirin. Either treatment of ibuprofen or aspirin, that showed no effects on METH-induced hyperthermia, significantly blocked the METH-induced striatal cyclooxygenase (COX) expression. Furthermore, the treatment of an intrinsic PPAR gamma ligand 15d-PG J2 also attenuated METH injections-induced reduction of striatal DAT. Therefore, the present study suggests the involvement of reduction of PPAR gamma expression in METH-induced neurotoxicity. Taken together with the previous report showing protective effects of other PPAR gamma ligand, these results imply that the protective effects of ibuprofen against METH-induced neurotoxicity may be based, in part, on its anti-inflammatory PPAR gamma agonistic properties, but not on its COX-inhibiting property or hypothermic effect.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Estimulantes del Sistema Nervioso Central/toxicidad , Cuerpo Estriado/efectos de los fármacos , Ibuprofeno/farmacología , Metanfetamina/toxicidad , Fármacos Neuroprotectores/farmacología , PPAR gamma/agonistas , Animales , Aspirina/farmacología , Temperatura Corporal/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Ciclooxigenasa 1/biosíntesis , Ciclooxigenasa 2/biosíntesis , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Fiebre/inducido químicamente , Fiebre/fisiopatología , Inmunohistoquímica , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Microglía/patología , PPAR gamma/biosíntesis , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología
19.
Neurotox Res ; 14(4): 295-305, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19073433

RESUMEN

Aggregation of proteins in the centrosome is implicated in the pathophysiology of Parkinson's disease. However, the relevance of the centrosome in neurodegeneration is still obscure. Centrosome duplication is initiated by the cyclin E/cyclin-dependent kinase 2 (Cdk2) complex. The present study determined changes in cyclin E or Cdk2 expression and in the centrosomal structure in dopaminergic neuronal CATH.a cells exposed to 50, 100 and 150 micromolar dopamine (DA) for 24 h. DA induced significant increase in Cdk2 protein and cyclin E protein, but not cyclin e mRNA. In DA-treated cells, the intense cyclin E- and Cdk2-immunofluorescence signals were co-localized around large and supernumerary centrosomes, and these two parameters of centrosome amplification were significantly increased compared with the control. Simultaneous co-treatment with DA and a Cdk2 inhibitor blocked centrosome amplification and enhanced cell viability. Our results demonstrated that DA could lead to cyclin E accumulation and Cdk2 up-regulation triggering supernumerary centrosomes and apoptotic cell death.


Asunto(s)
Muerte Celular/efectos de los fármacos , Centrosoma/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Dopamina/toxicidad , Neuronas/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Muerte Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Supervivencia Celular/efectos de los fármacos , Centrosoma/diagnóstico por imagen , Centrosoma/metabolismo , Centrosoma/fisiología , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Dopamina/genética , Indoles/farmacología , Ratones , Neuronas/ultraestructura , ARN Mensajero/metabolismo , Ultrasonografía , Regulación hacia Arriba
20.
Biochem Biophys Res Commun ; 377(4): 1051-6, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18955030

RESUMEN

Disrupted-In-Schizophrenia 1 (DISC1) was identified as a novel gene disrupted by a (1;11)(q42.1;q14.3) translocation segregating with schizophrenia, bipolar disorder and other major mental illnesses in a Scottish family. We previously identified 446-533 amino acids of DISC1 as the kendrin-binding region by means of a directed yeast two-hybrid interaction assay and showed that the DISC1-kendrin interaction is indispensable for the centrosomal localization of DISC1. In this study, to confirm the DISC1-kendrin interaction, we examined the interaction between deletion mutants of DISC1 and kendrin. Then, we demonstrated that the carboxy-terminus of DISC1 is indispensable for the interaction with kendrin. Furthermore, the immunocytochemistry revealed that the carboxy-terminus of DISC1 is also required for the centrosomal targeting of DISC1. Overexpression of the DISC1-binding region of kendrin or the DISC1 deletion mutant lacking the kendrin-binding region impairs the microtubule organization. These findings suggest that the DISC1-kendrin interaction plays a key role in the microtubule dynamics.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Centrosoma/metabolismo , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Células COS , Centrosoma/ultraestructura , Chlorocebus aethiops , Humanos , Inmunoprecipitación , Microtúbulos/ultraestructura , Proteínas del Tejido Nervioso/genética , Estructura Terciaria de Proteína , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA