Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35145028

RESUMEN

The cerebellum, the site where protein kinase C (PKC) was first discovered, contains the highest amount of PKC in the central nervous system, with PKCγ being the major isoform. Systemic PKCγ-knockout (KO) mice showed impaired motor coordination and deficient pruning of surplus climbing fibers (CFs) from developing cerebellar Purkinje cells (PCs). However, the physiological significance of PKCγ in the mature cerebellum and the cause of motor incoordination remain unknown. Using adeno-associated virus vectors targeting PCs, we showed that impaired motor coordination was restored by re-expression of PKCγ in mature PKCγ-KO mouse PCs in a kinase activity-dependent manner, while normal motor coordination in mature Prkcgfl/fl mice was impaired by the Cre-dependent removal of PKCγ from PCs. Notably, the rescue or removal of PKCγ from mature PKCγ-KO or Prkcgfl/fl mice, respectively, did not affect the CF innervation profile of PCs, suggesting the presence of a mechanism distinct from multiple CF innervation of PCs for the motor defects in PKCγ-deficient mice. We found marked potentiation of Ca2+-activated large-conductance K+ (BK) channel currents in PKCγ-deficient mice, as compared to wild-type mice, which decreased the membrane resistance, resulting in attenuation of the electrical signal during the propagation and significant alterations of the complex spike waveform. These changes in PKCγ-deficient mice were restored by the rescue of PKCγ or pharmacological suppression of BK channels. Our results suggest that PKCγ is a critical regulator that negatively modulates BK currents in PCs, which significantly influences PC output from the cerebellar cortex and, eventually, motor coordination.


Asunto(s)
Terapia Genética , Actividad Motora/genética , Canales de Potasio Calcio-Activados/metabolismo , Proteína Quinasa C/metabolismo , Células de Purkinje/enzimología , Animales , Señalización del Calcio , Eliminación de Gen , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Canales de Potasio Calcio-Activados/genética , Proteína Quinasa C/genética , Potenciales Sinápticos
2.
Int J Mol Sci ; 24(17)2023 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-37686396

RESUMEN

Activation of Gq-type G protein-coupled receptors (GPCRs) gives rise to large cytosolic Ca2+ elevations in astrocytes. Previous in vitro and in vivo studies have indicated that astrocytic Ca2+ elevations are closely associated with diameter changes in the nearby blood vessels, which astrocytes enwrap with their endfeet. However, the causal relationship between astrocytic Ca2+ elevations and blood vessel diameter changes has been questioned, as mice with diminished astrocytic Ca2+ signaling show normal sensory hyperemia. We addressed this controversy by imaging cortical vasculature while optogenetically elevating astrocyte Ca2+ in a novel transgenic mouse line, expressing Opto-Gq-type GPCR Optoα1AR (Astro-Optoα1AR) in astrocytes. Blue light illumination on the surface of the somatosensory cortex induced Ca2+ elevations in cortical astrocytes and their endfeet in mice under anesthesia. Blood vessel diameter did not change significantly with Optoα1AR-induced Ca2+ elevations in astrocytes, while it was increased by forelimb stimulation. Next, we labeled blood plasma with red fluorescence using AAV8-P3-Alb-mScarlet in Astro-Optoα1AR mice. We were able to identify arterioles that display diameter changes in superficial areas of the somatosensory cortex through the thinned skull. Photo-stimulation of astrocytes in the cortical area did not result in noticeable changes in the arteriole diameters compared with their background strain C57BL/6. Together, compelling evidence for astrocytic Gq pathway-induced vasodiameter changes was not observed. Our results support the notion that short-term (<10 s) hyperemia is not mediated by GPCR-induced astrocytic Ca2+ signaling.


Asunto(s)
Astrocitos , Hiperemia , Animales , Ratones , Ratones Endogámicos C57BL , Circulación Cerebrovascular , Transducción de Señal , Ratones Transgénicos
3.
Cerebellum ; 21(5): 776-783, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35218526

RESUMEN

The cerebellum contains the highest density of protein kinase C (PKC) in the central nervous system. PKCγ, the major isotype accounting for over half of the PKCs in the cerebellum, is expressed exclusively in Purkinje cells (PCs). Inactivated PKCγ, which is localized in the cytoplasm of PC dendrites and soma, begins to translocate to the cell membrane upon activation. However, the physiological conditions that induce PKCγ translocation in PC remain largely unknown. Here, we virally expressed PKCγ-GFP in PCs and examined the conditions that induced its translocation to PC dendrites by whole-cell patch clamp analysis combined with confocal GFP fluorescence imaging. A single or repetitive (150 pulses at 5 Hz for 30 s) electrical stimulation to a climbing fiber (CF), which produced a complex spike(s) in PC, failed to induce translocation of PKCγ-GFP to the dendritic shaft of PCs. Direct current injection (+ 2 nA for 3 s) to PC also did not induce the translocation, although PCs generated simple spikes continuously at high rates. In contrast, high-frequency parallel fiber (PF) stimulation (50 pulses at 50 Hz for 1 s), which triggered action potentials followed by sustained depolarization (known as mGluR1-mediated slow depolarization), caused translocation of cytoplasmic PKCγ-GFP to the membrane. Low-frequency PF stimulation (150 pulses at 5 Hz for 30 s) induced continuous simple spike firing but did not induce translocation. These results suggest that CF-triggered depolarization, which causes Ca2+ influx through voltage-gated Ca2+ channels throughout PC dendrites and somas, is insufficient to induce the translocation of PKCγ, instead requiring high-frequency PF stimulation that activates mGluR1.


Asunto(s)
Células de Purkinje , Sinapsis , Cerebelo/fisiología , Dendritas/metabolismo , Proteína Quinasa C/metabolismo , Células de Purkinje/fisiología , Sinapsis/fisiología
4.
J Pathol ; 255(4): 362-373, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34370295

RESUMEN

Urinary fatty acid binding protein 1 (FABP1, also known as liver-type FABP) has been implicated as a biomarker of acute kidney injury (AKI) in humans. However, the precise biological mechanisms underlying its elevation remain elusive. Here, we show that urinary FABP1 primarily reflects impaired protein reabsorption in proximal tubule epithelial cells (PTECs). Bilateral nephrectomy resulted in a marked increase in serum FABP1 levels, suggesting that the kidney is an essential organ for removing serum FABP1. Injected recombinant FABP1 was filtered through the glomeruli and robustly reabsorbed via the apical membrane of PTECs. Urinary FABP1 was significantly elevated in mice devoid of megalin, a giant endocytic receptor for protein reabsorption. Elevation of urinary FABP1 was also observed in patients with Dent disease, a rare genetic disease characterized by defective megalin function in PTECs. Urinary FABP1 levels were exponentially increased following acetaminophen overdose, with both nephrotoxicity and hepatotoxicity observed. FABP1-deficient mice with liver-specific overexpression of FABP1 showed a massive increase in urinary FABP1 levels upon acetaminophen injection, indicating that urinary FABP1 is liver-derived. Lastly, we employed transgenic mice expressing diphtheria toxin receptor (DT-R) either in a hepatocyte- or in a PTEC-specific manner, or both. Upon administration of diphtheria toxin (DT), massive excretion of urinary FABP1 was induced in mice with both kidney and liver injury, while mice with either injury type showed marginal excretion. Collectively, our data demonstrated that intact PTECs have a considerable capacity to reabsorb liver-derived FABP1 through a megalin-mediated mechanism. Thus, urinary FABP1, which is synergistically enhanced by concurrent liver injury, is a biomarker for impaired protein reabsorption in AKI. These findings address the use of urinary FABP1 as a biomarker of histologically injured PTECs that secrete FABP1 into primary urine, and suggest the use of this biomarker to simultaneously monitor impaired tubular reabsorption and liver function. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Lesión Renal Aguda , Biomarcadores/orina , Proteínas de Unión a Ácidos Grasos/orina , Hepatopatías , Animales , Humanos , Ratones
5.
Neuropathol Appl Neurobiol ; 47(2): 198-209, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32722888

RESUMEN

AIMS: Chaperone-mediated autophagy (CMA) is a pathway involved in the autophagy lysosome protein degradation system. CMA has attracted attention as a contributing factor to neurodegenerative diseases since it participates in the degradation of disease-causing proteins. We previously showed that CMA is generally impaired in cells expressing the proteins causing spinocerebellar ataxias (SCAs). Therefore, we investigated the effect of CMA impairment on motor function and the neural survival of cerebellar neurons using the micro RNA (miRNA)-mediated knockdown of lysosome-associated protein 2A (LAMP2A), a CMA-related protein. METHODS: We injected adeno-associated virus serotype 9 vectors, which express green fluorescent protein (GFP) and miRNA (negative control miRNA or LAMP2A miRNA) under neuron-specific synapsin I promoter, into cerebellar parenchyma of 4-week-old ICR mice. Motor function of mice was evaluated by beam walking and footprint tests. Immunofluorescence experiments of cerebellar slices were conducted to evaluate histological changes in cerebella. RESULTS: GFP and miRNA were expressed in interneurons (satellite cells and basket cells) in molecular layers and granule cells in the cerebellar cortices, but not in cerebellar Purkinje cells. LAMP2A knockdown in cerebellar neurons triggered progressive motor impairment, prominent loss of cerebellar Purkinje cells, interneurons, granule cells at the late stage, and astrogliosis and microgliosis from the early stage. CONCLUSIONS: CMA impairment in cerebellar interneurons and granule cells triggers the progressive ataxic phenotype, gliosis and the subsequent degeneration of cerebellar neurons, including Purkinje cells. Our present findings strongly suggest that CMA impairment is related to the pathogenesis of various SCAs.


Asunto(s)
Ataxia Cerebelosa/patología , Cerebelo/patología , Autofagia Mediada por Chaperones/fisiología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Degeneración Nerviosa/patología , Neuronas/patología , Animales , Ataxia Cerebelosa/metabolismo , Cerebelo/metabolismo , Ratones Endogámicos ICR , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Fenotipo
6.
J Neurosci ; 39(32): 6339-6353, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31201232

RESUMEN

ADP-ribosylation factors (ARFs) are a family of small monomeric GTPases comprising six members categorized into three classes: class I (ARF1, 2, and 3), class II (ARF4 and 5), and class III (ARF6). In contrast to class I and III ARFs, which are the key regulators in vesicular membrane trafficking, the cellular function of class II ARFs remains unclear. In the present study, we generated class II ARF-deficient mice and found that ARF4+/-/ARF5-/- mice exhibited essential tremor (ET)-like behaviors. In vivo electrophysiological recordings revealed that ARF4+/-/ARF5-/- mice of both sexes exhibited abnormal brain activity when moving, raising the possibility of abnormal cerebellar excitability. Slice patch-clamp experiments demonstrated the reduced excitability of the cerebellar Purkinje cells (PCs) in ARF4+/-/ARF5-/- mice. Immunohistochemical and electrophysiological analyses revealed a severe and selective decrease of pore-forming voltage-dependent Na+ channel subunit Nav1.6, important for maintaining repetitive action potential firing, in the axon initial segment (AIS) of PCs. Importantly, this decrease in Nav1.6 protein localized in the AIS and the consequent tremors in ARF4+/-/ARF5-/- mice could be alleviated by the PC-specific expression of ARF5 using adeno-associated virus vectors. Together, our data demonstrate that the decreased expression of the class II ARF proteins in ARF4+/-/ARF5-/- mice, leading to a haploinsufficiency of ARF4 in the absence of ARF5, impairs the localization of Nav1.6 to the AIS and hence reduces the membrane excitability in PCs, resulting in the ET-like movement disorder. We suggest that class II ARFs function in localizing specific proteins, such as Nav1.6, to the AIS.SIGNIFICANCE STATEMENT We found that decreasing the expression of class II ARF proteins, through the generation of ARF4+/-/ARF5-/- mice, impairs Nav1.6 distribution to the axon initial segment (AIS) of cerebellar Purkinje cells (PCs), thereby resulting in the impairment of action potential firing of PCs. The ARF4+/-/ARF5-/- mutant mice exhibited movement-associated essential tremor (ET)-like behavior with pharmacological profiles similar to those in ET patients. The exogenous expression of ARF5 reduced the tremor phenotype and restored the localization of Nav1.6 immunoreactivity to the AIS in ARF4+/-/ARF5-/- mice. Thus, our results suggest that class II ARFs are involved in the localization of Nav1.6 to the AISs in cerebellar PCs and that the reduction of class II ARF activity leads to ET-like movement disorder.


Asunto(s)
Factores de Ribosilacion-ADP/fisiología , Axones/metabolismo , Trastornos del Movimiento/etiología , Canal de Sodio Activado por Voltaje NAV1.6/fisiología , Células de Purkinje/metabolismo , Temblor/etiología , Factores de Ribosilacion-ADP/deficiencia , Factores de Ribosilacion-ADP/genética , Potenciales de Acción , Animales , Dependovirus/genética , Electroencefalografía , Electromiografía , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Genotipo , Movimientos de la Cabeza , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos del Movimiento/metabolismo , Trastornos del Movimiento/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.6/deficiencia , Técnicas de Placa-Clamp , Transporte de Proteínas , Células de Purkinje/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Método Simple Ciego , Temblor/metabolismo , Temblor/fisiopatología
7.
Biochem Biophys Res Commun ; 528(1): 199-205, 2020 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-32487317

RESUMEN

Glucocorticoids are released from the adrenal cortex and are important for regulating various physiological functions. However, a persistent increase in glucocorticoids due to chronic stress causes various dysfunctions in the central nervous system which can lead to mental disorders such as depression. Macroautophagy, one of the pathways of the autophagy-lysosome protein degradation system, is dysregulated in psychiatric disorders, implicating a disturbance of protein degradation in the pathogenesis of psychiatric disorders. In the present study, we investigated whether glucocorticoids affect the activity of chaperone-mediated autophagy (CMA) and microautophagy (mA), the other two pathways of the autophagy-lysosome system. Treatment of human-derived AD293 cells and primary cultured rat cortical neurons with dexamethasone, a potent glucocorticoid receptor agonist, and endogenous glucocorticoids decreased both CMA and mA activities. However, this decrease was significantly suppressed by treatment with RU-486, a glucocorticoid receptor antagonist. In addition, dexamethasone significantly decreased lysosomal Hsc70. These findings suggest that glucocorticoids negatively regulate CMA and mA in a glucocorticoid receptor-dependent manner, and provide evidence for CMA and mA as novel therapeutic targets for depression.


Asunto(s)
Autofagia Mediada por Chaperones/efectos de los fármacos , Glucocorticoides/farmacología , Microautofagia/efectos de los fármacos , Animales , Línea Celular , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratas
8.
Neurobiol Dis ; 121: 263-273, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30343032

RESUMEN

Cerebellar Purkinje cells (PCs) are the sole output neurons of the cerebellar cortex, and damage to PCs results in motor deficits. Spinocerebellar ataxia type 3 (SCA3, also known as Machado-Joseph disease), a hereditary neurodegenerative disease, is caused by an abnormal expansion of the polyglutamine tract in the causative ATXN3 protein. SCA3 affects a wide range of cells in the central nervous system, including those in the cerebellum. To unravel SCA3 pathology, we used adeno-associated virus serotype 9 (AAV9) vectors to express full-length ATXN3 with an abnormally expanded 89 polyglutamine stretch (ATXN3[Q89]) in cerebellar neurons of mature wild-type mice. Mice expressing ATXN3[Q89] exhibited motor impairment in a manner dependent on the viral titer. Immunohistochemistry of the cerebellum showed ubiquitinated nuclear aggregates in PCs; degeneration of PC dendrites; and a significant decrease in multiple proteins including retinoid-related orphan receptor α (RORα), a transcription factor, and type 1 metabotropic glutamate receptor (mGluR1) signaling molecules. Patch clamp analysis of ATXN3[Q89]-expressing PCs revealed marked defects in mGluR1 signaling. Notably, the emergence of behavioral, morphological, and functional defects was inhibited by a single injection of SR1078, an RORα/γ agonist. These results suggest that RORα plays a key role in mutant ATXN3-mediated aberrant phenotypes and that the pharmacological enhancement of RORα could function as a method for therapeutic intervention in SCA3.


Asunto(s)
Ataxina-3/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células de Purkinje/metabolismo , Proteínas Represoras/metabolismo , Animales , Ataxina-3/genética , Dendritas/patología , Humanos , Enfermedad de Machado-Joseph/patología , Ratones Endogámicos C57BL , Péptidos/genética , Agregación Patológica de Proteínas/metabolismo , Células de Purkinje/patología , Receptores de Glutamato Metabotrópico/metabolismo , Proteínas Represoras/genética , Transducción de Señal
9.
J Pharmacol Sci ; 140(2): 201-204, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31178328

RESUMEN

Autophagy-lysosome proteolysis is classified into macroautophagy (MA), microautophagy (mA) and chaperone-mediated autophagy (CMA). In contrast to MA and CMA, mA have been mainly studied in yeast. In 2011, mammalian mA was identified as a pathway to deliver cytosolic proteins into multivesicular bodies. However, its molecular mechanism is quite different from yeast mA. Using a cell-based method to evaluate mA and CMA, we revealed that rapamycin, an activator of yeast mA, significantly activated mammalian mA. Although rapamycin activates MA, mA was also activated by rapamycin in MA-deficient cells. These findings suggest that rapamycin is a first-identified activator of mammalian mA.


Asunto(s)
Microautofagia/efectos de los fármacos , Sirolimus/farmacología , Animales , Células Cultivadas , Autofagia Mediada por Chaperones/efectos de los fármacos , Ratones , Ratones Noqueados
10.
Mol Cell Neurosci ; 93: 36-47, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30342995

RESUMEN

Hydrogen sulfide and reactive sulfur species are regulators of physiological functions, have antioxidant effects against oxidative stresses, and are endogenously generated from l-cysteine. Recently, a novel pathway that generates hydrogen sulfide and reactive sulfur species from d-cysteine has been identified. d-Amino acid oxidase (DAO) is involved in this pathway and, among the various brain regions, is especially abundant in the cerebellum. d-Cysteine has been found to be a better substrate in the generation of hydrogen sulfide in the cerebellum than l-cysteine. Therefore, d-cysteine might be a novel neuroprotectant against cerebellar diseases such as spinocerebellar ataxia (SCA). However, it remains unknown if d-cysteine affects cerebellar Purkinje cells (PCs), which are important for cerebellar functions and are frequently degenerated in SCA patients. In the present study, we investigated whether the production of hydrogen sulfide from d-cysteine affects the dendritic development of cultured PCs. d-Cysteine was found to enhance the dendritic development of PCs significantly, while l-cysteine impaired it. The effect of d-cysteine was inhibited by simultaneous treatment with DAO inhibitors and was reproduced by treatment with 3-mercaptopyruvate, a metabolite of d-cysteine produced by the action of DAO, and disodium sulfide, a donor of hydrogen sulfide. In addition, hydrogen sulfide was immediately produced in cerebellar primary cultures after treatment with d-cysteine and 3-mercaptopyruvate. These findings suggest that d-cysteine enhances the dendritic development of primary cultured PCs via the generation of hydrogen sulfide.


Asunto(s)
Cisteína/farmacología , Sulfuro de Hidrógeno/metabolismo , Neurogénesis/efectos de los fármacos , Células de Purkinje/efectos de los fármacos , Animales , Antioxidantes/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cisteína/análogos & derivados , Cisteína/metabolismo , Estrés Oxidativo/efectos de los fármacos , Células de Purkinje/metabolismo , Ratas
11.
Neurobiol Dis ; 120: 34-50, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30184469

RESUMEN

Spinocerebellar ataxia type 21 (SCA21) is caused by missense or nonsense mutations of the transmembrane protein 240 (TMEM240). Molecular mechanisms of SCA21 pathogenesis remain unknown because the functions of TMEM240 have not been elucidated. We aimed to reveal the molecular pathogenesis of SCA21 using cell and mouse models that overexpressed the wild-type and SCA21 mutant TMEM240. In HeLa cells, overexpressed TMEM240 localized around large cytoplasmic vesicles. The SCA21 mutation did not affect this localization. Because these vesicles contained endosomal markers, we evaluated the effect of TMEM240 fused with a FLAG tag (TMEM-FL) on endocytosis and autophagic protein degradation. Wild-type TMEM-FL significantly impaired clathrin-mediated endocytosis, whereas the SCA21 mutants did not. The SCA21 mutant TMEM-FL significantly impaired autophagic lysosomal protein degradation, in contrast to wild-type. Next, we investigated how TMEM240 affects the neural morphology of primary cultured cerebellar Purkinje cells (PCs). The SCA21 mutant TMEM-FL significantly prevented the dendritic development of PCs, in contrast to the wild-type. Finally, we assessed mice that expressed wild-type or SCA21 mutant TMEM-FL in cerebellar neurons using adeno-associated viral vectors. Mice expressing the SCA21 mutant TMEM-FL showed impaired motor coordination. Although the SCA21 mutant TMEM-FL did not trigger neurodegeneration, activation of microglia and astrocytes was induced before motor miscoordination. In addition, immunoblot experiments revealed that autophagic lysosomal protein degradation, especially chaperone-mediated autophagy, was also impaired in the cerebella that expressed the SCA21 mutant TMEM-FL. These dysregulated functions in vitro, and induction of early gliosis and lysosomal impairment in vivo by the SCA21 mutant TMEM240 may contribute to the pathogenesis of SCA21.


Asunto(s)
Lisosomas/metabolismo , Proteínas de la Membrana/biosíntesis , Mutación/fisiología , Neuroglía/metabolismo , Degeneraciones Espinocerebelosas/metabolismo , Animales , Femenino , Células HeLa , Humanos , Lisosomas/genética , Lisosomas/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroglía/patología , Embarazo , Ratas , Ratas Wistar , Degeneraciones Espinocerebelosas/genética
12.
Biochem J ; 474(12): 1993-2007, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28432259

RESUMEN

ATP-binding cassette (ABC) transporters are ubiquitously present in prokaryotic and eukaryotic cells. Binding of ATP to the nucleotide-binding domains (NBDs) elicits major conformational changes of the transporters resulting in the transport of the substrate across the membrane. The availability of a crystal structure of the NBDs enabled us to elucidate the local structure and small-scale dynamics in the NBDs. Here, we labeled the ABC transporter MsbA, a homodimeric flippase from Escherichia coli, with a fluorescent probe, Alexa532, within the NBDs. ATP application elicited collisional quenching, whereas no quenching was observed after the addition of ATP analogs or ATP hydrolysis inhibitors. The Alexa532-conjugated MsbA variants exhibited transition metal ion Förster resonance energy transfer (tmFRET) after the addition of Ni2+, and ATP decreased this Ni2+-mediated FRET of the NBDs. Structure modeling developed from crystallographic data and examination of tmFRET measurements of MsbA variants in the absence of ATP revealed the presence of metal ion-associated pockets (MiAPs) in the NBDs. Three histidines were predicted to participate in chelating Ni2+ in the two possible MiAPs. Performing histidine-substitution experiments with the NBDs showed that the dissociation constant for Ni2+ of MiAP2 was smaller than that of MiAP1. The structural allocation of the MiAPs was further supported by showing that the addition of Cu2+ resulted in higher quenching than Ni2+ Taken together, the present study showed that the NBDs contain two native binding sites for metal ions and ATP addition affects the Ni2+-binding activity of the MiAPs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Níquel/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Sustitución de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Cobre/metabolismo , Bases de Datos de Proteínas , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Histidina/química , Histidina/metabolismo , Cinética , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Homología Estructural de Proteína
13.
Glia ; 65(6): 974-989, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28295574

RESUMEN

Glial development is critical for the function of the central nervous system. CD38 is a multifunctional molecule with ADP-ribosyl cyclase activity. While critical roles of CD38 in the adult brain such as oxytocin release and social behavior have been reported, those in the developing brain remain largely unknown. Here we demonstrate that deletion of Cd38 leads to impaired development of astrocytes and oligodendrocytes in mice. CD38 is highly expressed in the developing brains between postnatal day 14 (P14) and day 28 (P28). In situ hybridization and FACS analysis revealed that CD38 is expressed predominantly in astrocytes in these periods. Analyses of the cortex of Cd38 knockout (Cd38-/- ) mice revealed delayed development of astrocytes and subsequently delayed differentiation of oligodendrocytes (OLs) at postnatal stages. In vitro experiments using primary OL cultures, mixed glial cultures, and astrocytic conditioned medium showed that astrocytic CD38 regulates the development of astrocytes in a cell-autonomous manner and the differentiation of OLs in a non-cell-autonomous manner. Further experiments revealed that connexin43 (Cx43) in astrocytes plays a promotive role for CD38-mediated OL differentiation. Finally, increased levels of NAD+ , caused by CD38 deficiency, are likely to be responsible for the suppression of astrocytic Cx43 expression and OL differentiation. Our data indicate that CD38 is a positive regulator of astrocyte and OL development.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , ADP-Ribosil Ciclasa/metabolismo , Astrocitos/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Oligodendroglía/metabolismo , ADP-Ribosil Ciclasa/genética , ADP-Ribosil Ciclasa 1/genética , Animales , Astrocitos/citología , Encéfalo/citología , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Conexina 43/metabolismo , Femenino , Masculino , Glicoproteínas de Membrana/genética , Ratones Endogámicos ICR , Ratones Noqueados , NAD/metabolismo , Oligodendroglía/citología , Ratas Wistar
14.
J Neurochem ; 143(6): 660-670, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29049849

RESUMEN

Cerebellar Purkinje cells (PCs) express two members of the classical protein kinase C (cPKC) subfamily, namely, PKCα and PKCγ. Previous studies on PKCγ knockout (KO) mice have revealed a critical role of PKCγ in the pruning of climbing fibers (CFs) from PCs during development. The question remains as to why only PKCγ and not PKCα is involved in CF synapse elimination from PCs. To address this question, we assessed the expression levels of PKCγ and PKCα in wild-type (WT) and PKCγ KO PCs using PC-specific quantitative real-time reverse transcription-polymerase chain reaction, western blotting, and immunohistochemical analysis. The results revealed that the vast majority of cPKCs in PCs were PKCγ, whereas PKCα accounted for the remaining minimal fraction. The amount of PKCα was not up-regulated in PKCγ KO PCs. Lentiviral expression of PKCα in PKCγ KO PCs resulted in a 10-times increase in the amount of PKCα mRNA in the PKCγ KO PCs, compared to that in WT PCs. Our quantification showed that the expression levels of cPKC mRNA in PKCγ KO PCs increased roughly from 1% to 22% of that in WT PCs solely through PKCα expression. The up-regulation of PKCα in PKCγ KO PCs significantly rescued the impaired CF synapse elimination. Although both PKCα and PKCγ are capable of pruning supernumerary CF synapses from developing PCs, these results suggest that the expression levels of cPKCs in PKCγ KO PCs are too low for CF pruning.


Asunto(s)
Cerebelo/enzimología , Cerebelo/crecimiento & desarrollo , Proteína Quinasa C/biosíntesis , Células de Purkinje/citología , Animales , Animales Recién Nacidos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Nerviosas/metabolismo , Isoformas de Proteínas , Transcriptoma
15.
Genes Cells ; 21(8): 861-73, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27377049

RESUMEN

The autophagy-lysosome protein degradation is further classified into macroautophagy (MA), microautophagy (mA), and chaperone-mediated autophagy (CMA). While MA is involved in various functions and disease pathogenesis, little is known about CMA and mA because of the absence of easy methods to assess their activities. We have recently established a method to assess CMA activity using glyceraldehyde 3-phosphate dehydrogenase (GAPDH), a CMA substrate, and HaloTag (HT) system. Another group has recently identified a mammalian mA pathway, in which substrates are delivered to late endosomes in an heat shock cognate protein (Hsc)70-dependent manner. Because Hsc70 is also involved in CMA, our method would detect both CMA and mA activities. In this study, we attempted to assess CMA and mA activities separately through the siRNA-mediated knockdown of CMA- and mA-related proteins. Knockdown of LAMP2A, a CMA-related protein, and TSG101, an mA-related protein, significantly but only partially decreased the punctate accumulation of GAPDH-HT in AD293 cells and primary cultured rat cortical neurons. Compounds that activate CMA significantly increased GAPDH-HT puncta in TSG101-knockdown cells, but not in LAMP2A-knockdown cells, suggesting that punctate accumulation of GAPDH-HT under LAMP2A- and TSG101-knockdown represents mA and CMA activities, respectively. We succeeded in establishing the method to separately evaluate CMA and mA activities by fluorescence observation.


Asunto(s)
Autofagia/genética , Proteínas de Unión al ADN/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Gliceraldehído 3-Fosfato Deshidrogenasa (NADP+)/biosíntesis , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Neoplasias/genética , Factores de Transcripción/genética , Animales , Endosomas/metabolismo , Gliceraldehído 3-Fosfato Deshidrogenasa (NADP+)/genética , Células HEK293 , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/aislamiento & purificación , Neuronas/metabolismo , Proteolisis , Ratas
16.
Genes Cells ; 21(7): 717-27, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27184081

RESUMEN

The identification of novel synthetic ligands for G protein-coupled receptors (GPCRs) is important not only for understanding human physiology, but also for the development of novel drugs, especially for orphan GPCRs for which endogenous ligands are unknown. One of the orphan GPCR subfamilies, Super conserved Receptor Expressed in Brain (SREB), consists of GPR27, GPR85 and GPR173 and is expressed in the central nervous system. We report herein the identification of inverse agonists for the SREB family without their agonists. We carried out an in vitro screening of 5472 chemical compounds from the RIKEN NPDepo chemical library. The binding of [(35) S]GTPγS to the GPR173-Gsα fusion protein expressed in Sf9 cells was measured and resulted in the identification of 8 novel GPR173 inverse agonists. The most potent compound showed an IC50 of approximately 8 µm. The identified compounds were also antagonists for other SREB members, GPR27 and GPR85. These results indicated that the SREB family could couple Gs-type G proteins, and SREB-Gsα fusion proteins showed significant constitutive activities. Moreover, a molecular model of GPR173 was constructed using the screening results. The combination of computational and biological methods will provide a unique approach to ligand identification for orphan GPCRs and brain research.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Proteínas del Tejido Nervioso/química , Receptores Acoplados a Proteínas G/química , Bibliotecas de Moléculas Pequeñas/química , Encéfalo/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Expresión Génica , Humanos , Ligandos , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Interfaz Usuario-Computador
17.
Cerebellum ; 16(5-6): 913-922, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28508170

RESUMEN

We investigated the neuron-specific enolase (NSE) promoter in terms of its promoter strength and neuronal specificity in the cerebellum in vivo. The 1.8 kb rat NSE promoter was divided into three regions, A (0.8 kb), B (0.7 kb), and C (0.3 kb), starting from the 5' side. Then, we made various deletion constructs and assessed them by virally expressing GFP under the control of one of the deleted promoters. Removing region A reduced GFP expression to ~6% of that of the original 1.8 kb promoter. Further deletion of region B (presence of region C alone) did not influence the promoter strength, but removing region B from the original 1.8 kb promoter reduced the GFP expression to ~6% of the original level, similar to the level observed after deletion of region A. Immunohistochemistry showed robust GFP expression in Purkinje cells and modest expression in interneurons by the original promoter. Removing region A and/or region B abolished the GFP expression in Purkinje cells in most cerebellar lobules, with the expression in interneurons almost unchanged. These results suggest that region C, which is a proximal 0.3 kb sequence, contains cis-acting elements that drive transcription predominantly in interneurons. The addition of either region A or B onto region C does not alter the promoter properties; however, the addition of both regions A and B to region C drastically enhanced the promoter activity in Purkinje cells, suggesting the synergistic action of cis-acting regulatory elements in regions A and B for strong activation in Purkinje cells.


Asunto(s)
Cerebelo/metabolismo , Neuronas/metabolismo , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Regiones Promotoras Genéticas , Animales , Cerebelo/citología , Dependovirus/genética , Regulación de la Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Ratones Endogámicos C57BL , Neuronas/citología , Ratas
19.
Cerebellum ; 13(1): 29-41, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23955261

RESUMEN

Spinocerebellar ataxia type 3 (SCA3) is caused by the abnormal expansion of CAG repeats within the ataxin-3 gene. Previously, we generated transgenic mice (SCA3 mice) that express a truncated form of ataxin-3 containing abnormally expanded CAG repeats specifically in cerebellar Purkinje cells (PCs). Here, we further characterize these SCA3 mice. Whole-cell patch-clamp analysis of PCs from advanced-stage SCA3 mice revealed a significant decrease in membrane capacitance due to poor dendritic arborization and the complete absence of metabotropic glutamate receptor subtype1 (mGluR1)-mediated retrograde suppression of synaptic transmission at parallel fiber terminals, with an overall preservation of AMPA receptor-mediated fast synaptic transmission. Because these cerebellar phenotypes are reminiscent of retinoic acid receptor-related orphan receptor α (RORα)-defective staggerer mice, we examined the levels of RORα in the SCA3 mouse cerebellum by immunohistochemistry and found a marked reduction of RORα in the nuclei of SCA3 mouse PCs. To confirm that the defects in SCA3 mice were caused by postnatal deposition of mutant ataxin-3 in PCs, not by genome disruption via transgene insertion, we tried to reduce the accumulation of mutant ataxin-3 in developing PCs by viral vector-mediated expression of CRAG, a molecule that facilitates the degradation of stress proteins. Concomitant with the removal of mutant ataxin-3, CRAG-expressing PCs had greater numbers of differentiated dendrites compared to non-transduced PCs and exhibited retrograde suppression of synaptic transmission following mGluR1 activation. These results suggest that postnatal nuclear accumulation of mutant ataxin-3 disrupts dendritic differentiation and mGluR-signaling in SCA3 mouse PCs, and this disruption may be caused by a defect in a RORα-driven transcription pathway.


Asunto(s)
Cerebelo/fisiología , Dendritas/fisiología , Proteínas Nucleares/metabolismo , Células de Purkinje/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Factores de Transcripción/metabolismo , Potenciales de Acción , Animales , Ataxina-3 , Núcleo Celular/fisiología , Cerebelo/crecimiento & desarrollo , Dendritas/patología , Capacidad Eléctrica , Técnicas In Vitro , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/patología , Enfermedad de Machado-Joseph/fisiopatología , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Péptidos , Células de Purkinje/patología , Receptores AMPA/metabolismo , Transmisión Sináptica , Factores de Transcripción/química , Factores de Transcripción/genética
20.
Neurosci Res ; 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38311032

RESUMEN

The potential role of astrocytes in lateral habenula (LHb) in modulating anxiety was explored in this study. The habenula are a pair of small nuclei located above the thalamus, known for their involvement in punishment avoidance and anxiety. Herein, we observed an increase in theta-band oscillations of local field potentials (LFPs) in the LHb when mice were exposed to anxiety-inducing environments. Electrical stimulation of LHb at theta-band frequency promoted anxiety-like behavior. Calcium (Ca2+) levels and pH in the cytosol of astrocytes and local brain blood volume changes were studied in mice expressing either a Ca2+ or a pH sensor protein specifically in astrocytes and mScarlet fluorescent protein in the blood plasma using fiber photometry. An acidification response to anxiety was observed. Photoactivation of archaerhopsin-T (ArchT), an optogenetic tool that acts as an outward proton pump, results in intracellular alkalinization. Photostimulation of LHb in astrocyte-specific ArchT-expressing mice resulted in dissipation of theta-band LFP oscillation in an anxiogenic environment and suppression of anxiety-like behavior. These findings provide evidence that LHb astrocytes modulate anxiety and may offer a new target for treatment of anxiety disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA