Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Neurosci ; 41(9): 2053-2068, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33478986

RESUMEN

Spinocerebellar ataxias (SCAs) are diseases characterized by cerebellar atrophy and loss of Purkinje neurons caused by mutations in diverse genes. In SCA14, the disease is caused by point mutations or small deletions in protein kinase C γ (PKCγ), a crucial signaling protein in Purkinje cells. It is still unclear whether increased or decreased PKCγ activity may be involved in the SCA14 pathogenesis. In this study, we present a new knock-in mouse model related to SCA14 with a point mutation in the pseudosubstrate domain, PKCγ-A24E, known to induce a constitutive PKCγ activation. In this protein conformation, the kinase domain of PKCγ is activated, but at the same time the protein is subject to dephosphorylation and protein degradation. As a result, we find a dramatic reduction of PKCγ protein expression in PKCγ-A24E mice of either sex. Despite this reduction, there is clear evidence for an increased PKC activity in Purkinje cells from PKCγ-A24E mice. Purkinje cells derived from PKCγ-A24E have short thickened dendrites typical for PKC activation. These mice also develop a marked ataxia and signs of Purkinje cell dysfunction making them an interesting new mouse model related to SCA. Recently, a similar mutation in a human patient was discovered and found to be associated with overt SCA14. RNA profiling of PKCγ-A24E mice showed a dysregulation of related signaling pathways, such as mGluR1 or mTOR. Our results show that the induction of PKCγ activation in Purkinje cells results in the SCA-like phenotype indicating PKC activation as one pathogenetic avenue leading to a SCA.SIGNIFICANCE STATEMENT Spinocerebellar ataxias (SCAs) are hereditary diseases affecting cerebellar Purkinje cells and are a one of neurodegenerative diseases. While mutation in several genes have been identified as causing SCAs, it is unclear how these mutations cause the disease phenotype. Mutations in PKCγ cause one subtype of SCAs, SCA14. In this study, we have generated a knock-in mouse with a mutation in the pseudosubstrate domain of PKCγ, which keeps PKCγ in the constitutive active open conformation. We show that this mutation leading to a constant activation of PKCγ results in a SCA-like phenotype in these mice. Our findings establish the constant activation of PKC signaling as one pathogenetic avenue leading to an SCA phenotype and a mechanism causing a neurodegenerative disease.


Asunto(s)
Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/genética , Animales , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Actividad Motora/fisiología , Mutación , Células de Purkinje/patología , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología
2.
Int J Mol Sci ; 23(16)2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-36012439

RESUMEN

Spinocerebellar ataxias (SCAs) are a heterogeneous group of autosomal dominantly inherited progressive disorders with degeneration and dysfunction of the cerebellum. Although different subtypes of SCAs are classified according to the disease-associated causative genes, the clinical syndrome of the ataxia is shared, pointing towards a possible convergent pathogenic pathway among SCAs. In this review, we summarize the role of SCA-associated gene function during cerebellar Purkinje cell development and discuss the relationship between SCA pathogenesis and neurodevelopment. We will summarize recent studies on molecules involved in SCA pathogenesis and will focus on the mGluR1-PKCγ signaling pathway evaluating the possibility that this might be a common pathway which contributes to these diseases.


Asunto(s)
Proteína Quinasa C , Receptores de Glutamato Metabotrópico , Ataxias Espinocerebelosas , Humanos , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Células de Purkinje/metabolismo , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal , Ataxias Espinocerebelosas/patología
3.
Eur J Neurosci ; 54(7): 6673-6684, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34536317

RESUMEN

Serine/threonine kinase 17b (STK17B, also known as DRAK2) is known to be a downstream effector of protein kinase C (PKC) in the immune system, in particular T lymphocytes. PKC activity also plays a critical role for dendritic development and synaptic maturation and plasticity in cerebellar Purkinje cells. We present evidence that STK17B is strongly expressed in mouse cerebellar Purkinje cells starting in the early postnatal period and remaining highly expressed throughout adult stages and that STK17B is a target of PKC phosphorylation in the cerebellum. STK17B overexpression potentiates the morphological changes of Purkinje cells seen after PKC activation, suggesting that it is a downstream effector of PKC. A phosphorylation mimetic STK17B variant induced a marked reduction of Purkinje cell dendritic tree size, whereas the inhibition of STK17B with the novel compound 16 (Cpd16) could partially rescue the morphological changes of the Purkinje cell dendritic tree after PKC activation. These findings show that STK17B signalling is an important downstream effector of PKC activation in Purkinje cells. Furthermore, STK17B was identified as a molecule being transcriptionally downregulated in mouse models of SCA1, SCA7, SCA14 and SCA41. The reduced expression of STK17B in these mouse models might protect Purkinje cell dendrites from the negative effects of overactivated PKC signalling. Our findings provide new insights in the role of STK17B for Purkinje cell dendritic development and the pathology of SCAs.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas Serina-Treonina Quinasas/genética , Células de Purkinje , Ataxias Espinocerebelosas , Animales , Cerebelo/metabolismo , Ratones , Proteína Quinasa C/metabolismo , Células de Purkinje/metabolismo , Serina , Ataxias Espinocerebelosas/genética
4.
Eur J Neurosci ; 46(5): 2108-2120, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28715135

RESUMEN

The Na+ /Ca2+ exchanger (NCX) is a bidirectional plasma membrane antiporter involved in Ca2+ homeostasis in eukaryotes. NCX has three isoforms, NCX1-3, and all of them are expressed in the cerebellum. Immunostaining on cerebellar slice cultures indicates that NCX is widely expressed in the cerebellum, including expression in Purkinje cells. The pharmacological blockade of the forward mode of NCX (Ca2+ efflux mode) by bepridil moderately inhibited growth and development of Purkinje cell dendritic arbor in cerebellar slice cultures. However, the blockade of the reverse mode (Ca2+ influx mode) by KB-R7943 severely reduced the dendritic arbor and induced a morphological change with thickened distal dendrites. The effect of KB-R7943 on dendritic growth was unrelated to the activity of voltage-gated calcium channels and was also apparent in the absence of bioelectrical activity indicating that it was mediated by NCX expressed in Purkinje cells. We have used additional NCX inhibitors including CB-DMB, ORM-10103, SEA0400, YM-244769, and SN-6 which have higher specificity for NCX isoforms and target either the forward, reverse, or both modes. These inhibitors caused a strong dendritic reduction similar to that seen with KB-R7943, but did not elicit thickening of distal dendrites. Our findings indicate that disturbance of the NCX-dependent calcium transport in Purkinje cells induces a reduction of dendritic arbor, which is presumably caused by changes in the calcium handling, and underline the importance of the calcium equilibrium for the dendritic development in cerebellar Purkinje cells.


Asunto(s)
Dendritas/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Células de Purkinje/efectos de los fármacos , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Animales , Bepridil/farmacología , Canales de Calcio/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Dendritas/metabolismo , Dendritas/patología , Inmunohistoquímica , Ratones , Microscopía Confocal , Plasticidad Neuronal/fisiología , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/patología , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Tiourea/análogos & derivados , Tiourea/farmacología , Técnicas de Cultivo de Tejidos
5.
FASEB J ; 30(2): 624-34, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26472337

RESUMEN

The cold-inducible RNA-binding motif protein 3 (RBM3) is involved in the protection of neurons in hypoxic-ischemic and neurodegenerative disorders. RBM3 belongs to a small group of proteins whose synthesis increases during hypothermia while global protein production is slowed down. To investigate the molecular mechanisms underlying RBM3 action, we subjected hippocampal organotypic slice cultures from RBM3 knockout mice to various stressors and found exuberant signaling of the endoplasmic reticulum (ER) stress pathway PRKR-like ER kinase (PERK)-eukaryotic translation initiation factor 2α (eIF2α)-CCAAT/enhancer-binding protein homologous protein (CHOP) as compared with wild-type mice. Further, blocking RBM3 expression in human embryonic kidney HEK293 cells by specific small interfering RNAs increased phosphorylation of PERK and eIF2α, whereas overexpression of RBM3 prevented PERK-eIF2α-CHOP signaling during ER stress induced by thapsigargin or tunicamycin. RBM3 did not affect expression of the ER stress sensor immunoglobulin binding protein/GRP78. However, based on affinity purification coupled with mass spectrometry, coimmunoprecipitation, and proximity ligation assay, we revealed that nuclear factor 90 (NF90) is a novel protein interactor of PERK and that this interaction is essential for RBM3-mediated regulation of PERK activity, which requires an RNA-dependent interaction. In conclusion, our data provide evidence for a central role of RBM3 in preventing cell death by inhibiting the PERK-eIF2α-CHOP ER stress pathway through cooperation with NF90.


Asunto(s)
Frío , Proteínas del Factor Nuclear 90/metabolismo , Proteínas de Unión al ARN/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Supervivencia Celular , Retículo Endoplásmico/fisiología , Chaperón BiP del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Ratones , Proteínas del Factor Nuclear 90/genética , Fosforilación , Proteínas de Unión al ARN/genética , Estrés Fisiológico/fisiología , Tapsigargina/farmacología , Técnicas de Cultivo de Tejidos , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/genética
6.
Glia ; 62(5): 829-39, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24550001

RESUMEN

Despite the vast abundance of glial progenitor cells in the mouse brain parenchyma, little is known about the molecular mechanisms driving their proliferation in the adult. Here we unravel a critical role of the G1 cell cycle regulator cyclin D1 in controlling cell division of glial cells in the cortical grey matter. We detect cyclin D1 expression in Olig2-immunopositive (Olig2+) oligodendrocyte progenitor cells, as well as in Iba1+ microglia and S100ß+ astrocytes in cortices of 3-month-old mice. Analysis of cyclin D1-deficient mice reveals a cell and stage-specific molecular control of cell cycle progression in the various glial lineages. While proliferation of fast dividing Olig2+ cells at early postnatal stages becomes gradually dependent on cyclin D1, this particular G1 regulator is strictly required for the slow divisions of Olig2+/NG2+ oligodendrocyte progenitors in the adult cerebral cortex. Further, we find that the population of mature oligodendrocytes is markedly reduced in the absence of cyclin D1, leading to a significant decrease in the number of myelinated axons in both the prefrontal cortex and the corpus callosum of 8-month-old mutant mice. In contrast, the pool of Iba1+ cells is diminished already at postnatal day 3 in the absence of cyclin D1, while the number of S100ß+ astrocytes remains unchanged in the mutant.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Ciclina D1/biosíntesis , Neuroglía/metabolismo , Células Madre/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , División Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Femenino , Masculino , Ratones , Ratones Noqueados
7.
Neurobiol Dis ; 70: 1-11, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24937631

RESUMEN

Spinocerebellar ataxias (SCAs) are hereditary diseases leading to Purkinje cell degeneration and cerebellar dysfunction. Most forms of SCA are caused by expansion of CAG repeats similar to other polyglutamine disorders such as Huntington's disease. In contrast, in the autosomal dominant SCA-14 the disease is caused by mutations in the protein kinase C gamma (PKCγ) gene which is a well characterized signaling molecule in cerebellar Purkinje cells. The study of SCA-14, therefore, offers the unique opportunity to reveal the molecular and pathological mechanism eventually leading to Purkinje cell dysfunction and degeneration. We have created a mouse model of SCA-14 in which PKCγ protein with a mutation found in SCA-14 is specifically expressed in cerebellar Purkinje cells. We find that in mice expressing the mutated PKCγ protein the morphology of Purkinje cells in cerebellar slice cultures is drastically altered and mimics closely the morphology seen after pharmacological PKC activation. Similar morphological abnormalities were seen in localized areas of the cerebellum of juvenile transgenic mice in vivo. In adult transgenic mice there is evidence for some localized loss of Purkinje cells but there is no overall cerebellar atrophy. Transgenic mice show a mild cerebellar ataxia revealed by testing on the rotarod and on the walking beam. Our findings provide evidence for both an increased PKCγ activity in Purkinje cells in vivo and for pathological changes typical for cerebellar disease thus linking the increased and dysregulated activity of PKCγ tightly to the development of cerebellar disease in SCA-14 and possibly also in other forms of SCA.


Asunto(s)
Proteína Quinasa C/metabolismo , Células de Purkinje/enzimología , Células de Purkinje/patología , Degeneraciones Espinocerebelosas/enzimología , Degeneraciones Espinocerebelosas/patología , Animales , Western Blotting , Cerebelo/enzimología , Cerebelo/crecimiento & desarrollo , Cerebelo/patología , Dendritas/enzimología , Dendritas/patología , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Ratones Transgénicos , Actividad Motora/fisiología , Mutación , Proteína Quinasa C/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Ataxias Espinocerebelosas , Técnicas de Cultivo de Tejidos
8.
Neural Plast ; 2013: 321685, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24288624

RESUMEN

Purkinje cells are the principal neurons of the cerebellar cortex and have an extensive and elaborate dendritic tree. Chronic activation of type I metabotropic glutamate receptors inhibits Purkinje cell dendritic growth in organotypic cerebellar slice cultures. This effect is mediated by calcium influx through P/Q-type and T-type Ca(2+) channels. We have now studied the role of the plasma membrane Ca(2+)-ATPase2 (PMCA2), a major calcium extrusion pump, for Purkinje cell dendritic development. We found that PMCA2 is strongly expressed in the plasma membrane and dendritic spines of Purkinje cells in organotypic slice cultures compatible with a role for controlling the local dendritic calcium equilibrium. Inhibition of PMCA2 activity by carboxyeosin resulted in a moderate reduction of Purkinje cell dendritic tree size indicating that the extrusion of calcium by PMCA2 is important for maintaining the dendritic calcium concentration and controlling dendritic growth. When inhibition of PMCA2 was combined with stimulation of type I metabotropic glutamate receptors, it partially rescued dendritic morphology. This protection can be explained by a compensatory inactivation of voltage-gated calcium channels in Purkinje cells after PMCA2 inhibition. Our results demonstrate that PMCA2 activity is an important regulator of the dendritic calcium equilibrium controlling Purkinje cell dendritic growth.


Asunto(s)
Membrana Celular/enzimología , Cerebelo/enzimología , Cerebelo/fisiología , Dendritas/enzimología , Dendritas/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/fisiología , Células de Purkinje/enzimología , Células de Purkinje/fisiología , Animales , Calcio/fisiología , Células Cultivadas , Inmunohistoquímica , Ratones , Ratones Endogámicos , Técnicas de Cultivo de Órganos , Receptores de Glutamato Metabotrópico/metabolismo
9.
Nat Genet ; 31(1): 47-54, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11967539

RESUMEN

Control of cellular survival and proliferation is dependent on extracellular signals and is a prerequisite for ordered tissue development and maintenance. Activation of the cAMP responsive element binding protein (CREB) by phosphorylation has been implicated in the survival of mammalian cells. To define its roles in the mouse central nervous system, we disrupted Creb1 in brain of developing and adult mice using the Cre/loxP system. Mice with a Crem(-/-) background and lacking Creb in the central nervous system during development show extensive apoptosis of postmitotic neurons. By contrast, mice in which both Creb1 and Crem are disrupted in the postnatal forebrain show progressive neurodegeneration in the hippocampus and in the dorsolateral striatum. The striatal phenotype is reminiscent of Huntington disease and is consistent with the postulated role of CREB-mediated signaling in polyglutamine-triggered diseases.


Asunto(s)
Encéfalo/fisiología , Degeneración Nerviosa/etiología , Proteínas Represoras , Factores de Transcripción/fisiología , Animales , Apoptosis , Cuerpo Estriado/patología , Modulador del Elemento de Respuesta al AMP Cíclico , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Femenino , Humanos , Enfermedad de Huntington/etiología , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Péptidos/genética , Fenotipo , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
10.
Front Mol Neurosci ; 16: 1182431, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37426070

RESUMEN

Spinocerebellar ataxias (SCAs) are a group of hereditary neurodegenerative diseases mostly affecting cerebellar Purkinje cells caused by a wide variety of different mutations. One subtype, SCA14, is caused by mutations of Protein Kinase C gamma (PKCγ), the dominant PKC isoform present in Purkinje cells. Mutations in the pathway in which PKCγ is active, i.e., in the regulation of calcium levels and calcium signaling in Purkinje cells, are the cause of several other variants of SCA. In SCA14, many of the observed mutations in the PKCγ gene were shown to increase the basal activity of PKCγ, raising the possibility that increased activity of PKCγ might be the cause of most forms of SCA14 and might also be involved in the pathogenesis of SCA in related subtypes. In this viewpoint and review article we will discuss the evidence for and against such a major role of PKCγ basal activity and will suggest a hypothesis of how PKCγ activity and the calcium signaling pathway may be involved in the pathogenesis of SCAs despite the different and sometimes opposing effects of mutations affecting these pathways. We will then widen the scope and propose a concept of SCA pathogenesis which is not primarily driven by cell death and loss of Purkinje cells but rather by dysfunction of Purkinje cells which are still present and alive in the cerebellum.

11.
Eur J Neurosci ; 35(1): 20-33, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22188405

RESUMEN

The development of a neuronal dendritic tree is modulated both by signals from afferent fibers and by an intrinsic program. We have previously shown that chronic activation of either type 1 metabotropic glutamate receptors (mGluR1s) or protein kinase C (PKC) in organotypic cerebellar slice cultures of mice and rats severely inhibits the growth and development of the Purkinje cell dendritic tree. The signaling events linking receptor activation to the regulation of dendritic growth remain largely unknown. We have studied whether channels allowing the entry of Ca(2+) into Purkinje cells, in particular the type 3 transient receptor potential cation channels (TRPC3s), P/Q-type Ca(2+) channels, and T-type Ca(2+) channels, might be involved in signaling after mGluR1 or PKC stimulation. We show that the inhibition of dendritic growth seen after mGluR1 or PKC stimulation is partially rescued by pharmacological blockade of P/Q-type and T-type Ca(2+) channels, indicating that activation of these channels mediating Ca(2+) influx contributes to the inhibition of dendritic growth. In contrast, the absence of Ca(2+) -permeable TRPC3s in TRPC3-deficient mice or pharmacological blockade had no effect on mGluR1-mediated and PKC-mediated inhibition of Purkinje cell dendritic growth. Similarly, blockade of Ca(2+) influx through glutamate receptor δ2 or R-type Ca(2+) channels or inhibition of release from intracellular stores did not influence mGluR1-mediated and PKC-mediated inhibition of Purkinje cell dendritic growth. These findings suggest that both T-type and P/Q-type Ca(2+) channels, but not TRPC3 or other Ca(2+) -permeable channels, are involved in mGluR1 and PKC signaling leading to the inhibition of dendritic growth in cerebellar Purkinje cells.


Asunto(s)
Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Canales de Calcio Tipo T/metabolismo , Dendritas/fisiología , Proteína Quinasa C/metabolismo , Células de Purkinje/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Calcio/metabolismo , Dendritas/efectos de los fármacos , Dendritas/ultraestructura , Activación Enzimática , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células de Purkinje/citología , Células de Purkinje/efectos de los fármacos , Ratas , Acetato de Tetradecanoilforbol/farmacología , Fosfolipasas de Tipo C/metabolismo
12.
Genes (Basel) ; 13(8)2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-36011327

RESUMEN

The autosomal dominant inherited spinocerebellar ataxias (SCAs) are a group of neurodegenerative disorders characterized by cerebellar atrophy and loss of Purkinje neurons. Spinocerebellar ataxia type 14 (SCA14) is a rare variant of SCAs caused by missense mutations or deletions in the PRKCG gene encoding the protein kinase C γ (PKCγ). Although mutated PKCγs are responsible for SCA14, it is still unclear exactly how mutated PKCγs are involved in SCA14 pathogenesis. Therefore, it is important to study how PKCγ signaling is altered in the cerebellum, which genes or signaling pathways are affected, and how this leads to neurological disease. In this study, we used a mouse line carrying a knock-in pseudo-substrate domain mutation in PKCγ (PKCγ-A24E) as an SCA14 model and performed RNA sequencing (RNA-seq) analysis at an early developmental timepoint (postnatal day 15) to investigate changes in the gene profile compared to wildtype mice. We analyzed both heterozygous (Het) PKCγ-A24E mice and homozygous (Homo) PKCγ-A24E mice for transcriptomic changes. The Het PKCγ-A24E mice reflects the situation observed in human SCA14 patient, while Homo PKCγ-A24E mice display stronger phenotypes with respect to Purkinje cell development and behavior. Our findings highlight an abundance of modifications affecting genes involved in developmental processes, suggesting that at least a part of the final phenotype is shaped by altered cerebellar development and is not only caused by changes in mature animals.


Asunto(s)
Ataxias Espinocerebelosas , Transcriptoma , Animales , Cerebelo/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Células de Purkinje/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
13.
Front Cell Dev Biol ; 10: 854273, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35712654

RESUMEN

CRISPR-Cas13 technology is rapidly evolving as it is a very specific tool for RNA editing and interference. Since there are no significant off-target effects via the Cas13-mediated method, it is a promising tool for studying gene function in differentiating neurons. In this study, we designed two crRNA targeting regulator of G-protein signaling 8 (RGS8), which is a signaling molecule associated with spinocerebellar ataxias. Using CRISPR-Cas13 technology, we found that both of crRNAs could specifically achieve RGS8 knockdown. By observing and comparing the dendritic growth of Purkinje cells, we found that CRISPR-Cas13-mediated RGS8 knockdown did not significantly affect Purkinje cell dendritic development. We further tested the role of RGS8 by classical RNAi. Again, the results of the RNAi-mediated RGS8 knockdown showed that reduced RGS8 expression did not significantly affect the dendritic growth of Purkinje cells. This is the first example of CRISPR-Cas13-mediated gene function study in Purkinje cells and establishes CRISPR-Cas13-mediated knockdown as a reliable method for studying gene function in primary neurons.

14.
J Neurosci ; 30(4): 1385-94, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20107064

RESUMEN

GABA(B) receptors are the G-protein-coupled receptors for GABA, the main inhibitory neurotransmitter in the brain. Two receptor subtypes, GABA(B(1a,2)) and GABA(B(1b,2)), are formed by the assembly of GABA(B1a) and GABA(B1b) subunits with GABA(B2) subunits. The GABA(B1b) subunit is a shorter isoform of the GABA(B1a) subunit lacking two N-terminal protein interaction motifs, the sushi domains. Selectively GABA(B1a) protein traffics into the axons of glutamatergic neurons, whereas both the GABA(B1a) and GABA(B1b) proteins traffic into the dendrites. The mechanism(s) and targeting signal(s) responsible for the selective trafficking of GABA(B1a) protein into axons are unknown. Here, we provide evidence that the sushi domains are axonal targeting signals that redirect GABA(B1a) protein from its default dendritic localization to axons. Specifically, we show that mutations in the sushi domains preventing protein interactions preclude axonal localization of GABA(B1a). When fused to CD8alpha, the sushi domains polarize this uniformly distributed protein to axons. Likewise, when fused to mGluR1a the sushi domains redirect this somatodendritic protein to axons, showing that the sushi domains can override dendritic targeting information in a heterologous protein. Cell surface expression of the sushi domains is not required for axonal localization of GABA(B1a). Altogether, our findings are consistent with the sushi domains functioning as axonal targeting signals by interacting with axonally bound proteins along intracellular sorting pathways. Our data provide a mechanistic explanation for the selective trafficking of GABA(B(1a,2)) receptors into axons while at the same time identifying a well defined axonal delivery module that can be used as an experimental tool.


Asunto(s)
Transporte Axonal/fisiología , Axones/metabolismo , Hipocampo/metabolismo , Inhibición Neural/fisiología , Receptores de GABA-B/metabolismo , Transmisión Sináptica/fisiología , Animales , Axones/ultraestructura , Antígenos CD8/genética , Antígenos CD8/metabolismo , Polaridad Celular/fisiología , Células Cultivadas , Dendritas/metabolismo , Dendritas/ultraestructura , Hipocampo/ultraestructura , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación/genética , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína/genética , Subunidades de Proteína/metabolismo , Transporte de Proteínas/fisiología , Receptores de GABA/química , Receptores de GABA/genética , Receptores de GABA/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de GABA-B/química , Receptores de GABA-B/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología
15.
BMC Neurosci ; 12: 89, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21896184

RESUMEN

BACKGROUND: Little is known about expression and function of the somatostatinergic system in the mammalian cochlea. We have previously shown that somatostatin administration may have a protective effect on gentamicin-induced hair cell loss. In this study, we have analyzed the cochlear expression of somatostatin receptor 1 (SST1) and somatostatin receptor 2 (SST2) at both the mRNA and the protein level in wild-type mice, as well as in SST1 and SST2 knock-out (KO) mice and in cultivated neurosensory cells. RESULTS: We demonstrate that the somatostatin receptors SST1 and SST2 are specifically expressed in outer and inner hair cells (HCs) of the organ of Corti (OC), as well as in defined supporting cells. The expression of SST1 and SST2 receptors in cultivated P5 mouse OC explants was similar to their expression in inner and outer hair cells. Somatostatin itself was not expressed in the mammalian cochlea, suggesting that somatostatin reaches its receptors either through the blood-labyrinthine barrier from the systemic circulation or via the endolymphatic duct from the endolymphatic sac. We used mice with a deletion of either SST1 or SST2 to learn more about the regulation of SST1 and SST2 receptor expression. We demonstrate that in SST1 KO mice, SST2 was expressed in outer HCs and Deiters' cells, but not in pillar cells or inner HCs, as compared with wild-type mice. In contrast, in SST2 KO mice, the expression pattern of the SST1 receptor was not altered relative to wild-type mice. CONCLUSIONS: These findings reveal that somatostatin receptors demonstrate specific expression in HCs and supporting cells of the mouse cochlea, and that absence of SST1 alters the expression of SST2. This specific expression pattern suggests that somatostatin receptors may have important functional roles in the inner ear.


Asunto(s)
Cóclea/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Tisular
16.
Toxins (Basel) ; 13(4)2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33916905

RESUMEN

The CRISPR-Cas13 system based on a bacterial enzyme has been explored as a powerful new method for RNA manipulation. Due to the high efficiency and specificity of RNA editing/interference achieved by this system, it is currently being developed as a new therapeutic tool for the treatment of neurological and other diseases. However, the safety of this new generation of RNA therapies is still unclear. In this study, we constructed a vector expressing CRISPR-Cas13 under a constitutive neuron-specific promoter. CRISPR-Cas13 from Leptotrichia wadei was expressed in primary cultures of mouse cortical neurons. We found that the presence of CRISPR-Cas13 impedes the development of cultured neurons. These results show a neurotoxic action of Cas13 and call for more studies to test for and possibly mitigate the toxic effects of Cas13 enzymes in order to improve CRISPR-Cas13-based tools for RNA targeting.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Corteza Cerebral/enzimología , Leptotrichia/enzimología , Proyección Neuronal , Neuronas/enzimología , Animales , Proteínas Asociadas a CRISPR/genética , Células Cultivadas , Corteza Cerebral/patología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Leptotrichia/genética , Ratones , Neuronas/patología
17.
Brain Sci ; 11(8)2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34439673

RESUMEN

RNA therapies using RNA editing and interference are currently being developed for neurological diseases. The CRISPR-Cas13 system, based on bacterial enzymes, holds great promise for developing efficient tools for RNA therapies. However, neurotoxic activity has been reported for Cas13a, and recent studies have reported toxic effects of PspCas13b and RfxCas13d during zebrafish and Drosophila embryonic development. It is important to investigate the safety of these bacterial enzymes in the context of the nervous system and neuronal development. In this study, we used mouse cerebellar Purkinje cells as a complex neuron type to test for the potential neurotoxic actions of RfxCas13d and PspCas13b. We found that PspCas13b significantly impeded the dendritic development of cultured Purkinje cells, similar to the neurotoxic action of Cas13a. In contrast, RfxCas13d did not exhibit a significant inhibition of dendritic development. A similar trend was found for axonal outgrowth. These results suggest varying neurotoxic properties for different Cas13 ortholog enzymes. We call for more studies to investigate, and possibly mitigate, the neurotoxicity of Cas13 proteins in order to improve the safety of the CRISPR-Cas13 system for RNA therapies.

18.
Biochim Biophys Acta Gen Subj ; 1865(6): 129869, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33581252

RESUMEN

• Specific gene knockdown in cerebellar Purkinje cells is a major challenge because Purkinje cells only make up a small fraction of the cerebellum and off-target effects of shRNA occur. • Little is known about the application of CRISPR-Cas13 and the resulting protein expression levels in Purkinje cells, a type of developing postmitotic neuron. • We explored the possibility of a Cas13-mediated conditional knockdown system for cerebellar Purkinje cells. This system can achieve a suppression of the target proteins restricted to Purkinje cells in mixed cerebellar cultures.


Asunto(s)
Sistemas CRISPR-Cas , Cerebelo/patología , Silenciador del Gen , Proteína Quinasa C/antagonistas & inhibidores , Células de Purkinje/patología , Animales , Cerebelo/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa C/genética , Células de Purkinje/metabolismo
19.
J Neurosci Res ; 88(4): 774-83, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19798747

RESUMEN

Purkinje cell loss by excitotoxic damage is a typical finding in many cerebellar diseases. One important aspect of this high sensitivity of Purkinje cells to excitotoxic death might be the enormous size of their dendritic tree, with a high load of excitatory glutamate receptors. We have studied whether reduction in the size of the dendritic tree might confer resistance against excitotoxic death to Purkinje cells. We have grown Purkinje cells in organotypic cerebellar slice cultures under chronic activation of metabotropic glutamate receptors or of protein kinase C. Both treatments strongly reduced dendritic tree size. After this treatment, cells were exposed to the glutamate receptor agonist AMPA, which has a strong excitotoxic effect on Purkinje cells. We found that Purkinje cells with small dendritic trees were as sensitive to AMPA exposure as untreated control cells with large dendritic trees. Immunostaining against vesicular glutamate transporter 1 revealed that the small dendritic trees were densely covered by glutamatergic terminals. Our results indicate that the expansion of the dendritic tree and the total number of AMPA receptors per neuron do not play a major role in determining the susceptibility of Purkinje cells to excitotoxic death.


Asunto(s)
Dendritas/patología , Agonistas de Aminoácidos Excitadores/toxicidad , Células de Purkinje/citología , Células de Purkinje/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/toxicidad , Animales , Animales Recién Nacidos , Calbindinas , Muerte Celular/efectos de los fármacos , Cerebelo/citología , Dendritas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Proteína G de Unión al Calcio S100/metabolismo , Sinapsis/efectos de los fármacos , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
20.
Front Cell Dev Biol ; 8: 569889, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33553137

RESUMEN

Spinocerebellar ataxias (SCAs) are a group of hereditary neurodegenerative diseases which are caused by diverse genetic mutations in a variety of different genes. We have identified RGS8, a regulator of G-protein signaling, as one of the genes which are dysregulated in different mouse models of SCA (e.g., SCA1, SCA2, SCA7, and SCA14). In the moment, little is known about the role of RGS8 for pathogenesis of spinocerebellar ataxia. We have studied the expression of RGS8 in the cerebellum in more detail and show that it is specifically expressed in mouse cerebellar Purkinje cells. In a mouse model of SCA14 with increased PKCγ activity, RGS8 expression was also increased. RGS8 overexpression could partially counteract the negative effects of DHPG-induced mGluR1 signaling for the expansion of Purkinje cell dendrites. Our results suggest that the increased expression of RGS8 is an important mediator of mGluR1 pathway dysregulation in Purkinje cells. These findings provide new insights in the role of RGS8 and mGluR1 signaling in Purkinje cells and for the pathology of SCAs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA