Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 43(36): 6249-6267, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37558490

RESUMEN

The dopaminergic neuromodulator system is fundamental to brain functions. Abnormal dopamine (DA) pathway is implicated in psychiatric disorders, including schizophrenia (SZ) and autism spectrum disorder (ASD). Mutations in Cullin 3 (CUL3), a core component of the Cullin-RING ubiquitin E3 ligase complex, have been associated with SZ and ASD. However, little is known about the function and mechanism of CUL3 in the DA system. Here, we show that CUL3 is critical for the function of DA neurons and DA-relevant behaviors in male mice. CUL3-deficient mice exhibited hyperactive locomotion, deficits in working memory and sensorimotor gating, and increased sensitivity to psychostimulants. In addition, enhanced DA signaling and elevated excitability of the VTA DA neurons were observed in CUL3-deficient animals. Behavioral impairments were attenuated by dopamine D2 receptor antagonist haloperidol and chemogenetic inhibition of DA neurons. Furthermore, we identified HCN2, a hyperpolarization-activated and cyclic nucleotide-gated channel, as a potential target of CUL3 in DA neurons. Our study indicates that CUL3 controls DA neuronal activity by maintaining ion channel homeostasis and provides insight into the role of CUL3 in the pathogenesis of psychiatric disorders.SIGNIFICANCE STATEMENT This study provides evidence that Cullin 3 (CUL3), a core component of the Cullin-RING ubiquitin E3 ligase complex that has been associated with autism spectrum disorder and schizophrenia, controls the excitability of dopamine (DA) neurons in mice. Its DA-specific heterozygous deficiency increased spontaneous locomotion, impaired working memory and sensorimotor gating, and elevated response to psychostimulants. We showed that CUL3 deficiency increased the excitability of VTA DA neurons, and inhibiting D2 receptor or DA neuronal activity attenuated behavioral deficits of CUL3-deficient mice. We found HCN2, a hyperpolarization-activated channel, as a target of CUL3 in DA neurons. Our findings reveal CUL3's role in DA neurons and offer insights into the pathogenic mechanisms of autism spectrum disorder and schizophrenia.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Esquizofrenia , Animales , Masculino , Ratones , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Ubiquitinas/metabolismo , Ubiquitinas/farmacología , Área Tegmental Ventral
2.
Mol Psychiatry ; 28(3): 1027-1045, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-33990773

RESUMEN

Dopamine (DA) neurons in the ventral tegmental area (VTA) are critical to coping with stress. However, molecular mechanisms regulating their activity and stress-induced depression were not well understood. We found that the receptor tyrosine kinase ErbB4 in VTA was activated in stress-susceptible mice. Deleting ErbB4 in VTA or in DA neurons, or chemical genetic inhibition of ErbB4 kinase activity in VTA suppressed the development of chronic social defeat stress (CSDS)-induced depression-like behaviors. ErbB4 activation required the expression of NRG1 in the laterodorsal tegmentum (LDTg); LDTg-specific deletion of NRG1 inhibited depression-like behaviors. NRG1 and ErbB4 suppressed potassium currents of VTA DA neurons and increased their firing activity. Finally, we showed that acute inhibition of ErbB4 after stress attenuated DA neuron hyperactivity and expression of depression-like behaviors. Together, these observations demonstrate a critical role of NRG1-ErbB4 signaling in regulating depression-like behaviors and identify an unexpected mechanism by which the LDTg-VTA circuit regulates the activity of DA neurons.


Asunto(s)
Depresión , Área Tegmental Ventral , Ratones , Animales , Área Tegmental Ventral/metabolismo , Neuronas Dopaminérgicas/metabolismo , Transducción de Señal , Fosforilación , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo
3.
Neuron ; 110(14): 2315-2333.e6, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35561677

RESUMEN

Heat perception enables acute avoidance responses to prevent tissue damage and maintain body thermal homeostasis. Unlike other modalities, how heat signals are processed in the spinal cord remains unclear. By single-cell gene profiling, we identified ErbB4, a transmembrane tyrosine kinase, as a novel marker of heat-sensitive spinal neurons in mice. Ablating spinal ErbB4+ neurons attenuates heat sensation. These neurons receive monosynaptic inputs from TRPV1+ nociceptors and form excitatory synapses onto target neurons. Activation of ErbB4+ neurons enhances the heat response, while inhibition reduces the heat response. We showed that heat sensation is regulated by NRG1, an activator of ErbB4, and it involves dynamic activity of the tyrosine kinase that promotes glutamatergic transmission. Evidence indicates that the NRG1-ErbB4 signaling is also engaged in hypersensitivity of pathological pain. Together, these results identify a spinal neuron connection consisting of ErbB4+ neurons for heat sensation and reveal a regulatory mechanism by the NRG1-ErbB4 signaling.


Asunto(s)
Calor , Neurregulina-1 , Neuronas , Sensación Térmica , Animales , Ratones , Neurregulina-1/farmacología , Neuronas/fisiología , Receptor ErbB-4/genética
4.
Curr Biol ; 31(15): 3330-3342.e7, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34143959

RESUMEN

Dopamine (DA) transmission is critical to motivation, movement, and emotion. Unlike glutamatergic and GABAergic synapses, the development of DA synapses is less understood. We show that bassoon (BSN) clusters along DA axons in the core of nucleus accumbens (NAcc) were increased in neonatal stages and reduced afterward, suggesting DA synapse elimination. Remarkably, DA neuron-specific ablating neuregulin 3 (NRG3), a protein whose levels correlate with BSN clusters, increased the clusters and impaired DA release and behaviors related to DA transmission. An unbiased screen of transmembrane proteins with the extracellular domain (ECD) of NRG3 identified Caspr3 (contactin associate-like protein 3) as a binding partner. Caspr3 was enriched in striatal medium spiny neurons (MSNs). NRG3 and Caspr3 interact in trans, which was blocked by Caspr3-ECD. Caspr3 null mice displayed phenotypes similar to those in DAT-Nrg3f/f mice in DA axonal BSN clusters and DA transmission. Finally, in vivo disruption of the NRG3-Caspr3 interaction increased BSN clusters. Together, these results demonstrate that DA synapse development is controlled by trans interaction between NRG3 in DA neurons and Caspr3 in MSNs, identifying a novel pair of cell adhesion molecules for brain circuit wiring.


Asunto(s)
Cuerpo Estriado , Dopamina , Neuronas Dopaminérgicas/citología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Neurregulinas/fisiología , Animales , Cuerpo Estriado/citología , Ratones , Ratones Noqueados , Sinapsis
5.
J Clin Invest ; 131(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33651714

RESUMEN

The excitability of interneurons requires Nav1.1, the α subunit of the voltage-gated sodium channel. Nav1.1 deficiency and mutations reduce interneuron excitability, a major pathological mechanism for epilepsy syndromes. However, the regulatory mechanisms of Nav1.1 expression remain unclear. Here, we provide evidence that neddylation is critical to Nav1.1 stability. Mutant mice lacking Nae1, an obligatory component of the E1 ligase for neddylation, in parvalbumin-positive interneurons (PVINs) exhibited spontaneous epileptic seizures and premature death. Electrophysiological studies indicate that Nae1 deletion reduced PVIN excitability and GABA release and consequently increased the network excitability of pyramidal neurons (PyNs). Further analysis revealed a reduction in sodium-current density, not a change in channel property, in mutant PVINs and decreased Nav1.1 protein levels. These results suggest that insufficient neddylation in PVINs reduces Nav1.1 stability and thus the excitability of PVINs; the ensuing increased PyN activity causes seizures in mice. Consistently, Nav1.1 was found reduced by proteomic analysis that revealed abnormality in synapses and metabolic pathways. Our findings describe a role of neddylation in maintaining Nav1.1 stability for PVIN excitability and reveal what we believe is a new mechanism in the pathogenesis of epilepsy.


Asunto(s)
Potenciales de Acción , Epilepsia/metabolismo , Interneuronas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Convulsiones/metabolismo , Animales , Modelos Animales de Enfermedad , Interneuronas/patología , Ratones , Ratones Mutantes , Canal de Sodio Activado por Voltaje NAV1.1/genética , Estabilidad Proteica , Células Piramidales/metabolismo , Células Piramidales/patología , Convulsiones/genética , Enzimas Activadoras de Ubiquitina/deficiencia , Enzimas Activadoras de Ubiquitina/metabolismo
6.
J Neurosci ; 40(38): 7241-7254, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32847967

RESUMEN

Maladaptation to stress is a critical risk factor in stress-related disorders, such as major depression and post-traumatic stress disorder (PTSD). Dopamine signaling in the nucleus accumbens (NAc) has been shown to modulate behavior by reinforcing learning and evading aversive stimuli, which are important for the survival of animals under environmental challenges such as stress. However, the mechanisms through which dopaminergic transmission responds to stressful events and subsequently regulates its downstream neuronal activity during stress remain unknown. To investigate how dopamine signaling modulates stress-coping behavior, we measured the subsecond fluctuation of extracellular dopamine concentration and pH using fast scanning cyclic voltammetry (FSCV) in the NAc, a postsynaptic target of midbrain dopaminergic neurons, in male mice engaged in a tail suspension test (TST). The results revealed a transient decrease in dopamine concentration and an increase in pH levels when the animals changed behaviors, from being immobile to struggling. Interestingly, optogenetic inhibition of dopamine release in NAc, potentiated the struggling behavior in animals under the TST. We then addressed the causal relationship of such a dopaminergic transmission with behavioral alterations by knocking out both the dopamine receptors, i.e., D1 and D2, in the NAc using viral vector-mediated genome editing. Behavioral analyses revealed that male D1 knock-out mice showed significantly more struggling bouts and longer struggling durations during the TST, while male D2 knock-out mice did not. Our results therefore indicate that D1 dopaminergic signaling in the NAc plays a pivotal role in the modulation of stress-coping behaviors in animals under tail suspension stress.SIGNIFICANCE STATEMENT The tail suspension test (TST) has been widely used as a despair-based behavioral assessment to screen the antidepressant so long. Despite its prevalence in the animal studies, the neural substrate underlying the changes of behavior during the test remains unclear. This study provides an evidence for a role of dopaminergic transmission in the modulation of stress-coping behavior during the TST, a despair test widely used to screen the antidepressants in rodents. Taking into consideration the fact that the dopamine metabolism is upregulated by almost all antidepressants, a part of which acts directly on the dopaminergic transmission, current results would uncover the molecular mechanism through which the dopaminergic signaling mediates antidepressant effect with facilitation of the recovery from the despair-like behavior in the TST.


Asunto(s)
Adaptación Psicológica , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Núcleo Accumbens/metabolismo , Estrés Psicológico/metabolismo , Animales , Línea Celular Tumoral , Neuronas Dopaminérgicas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/citología , Núcleo Accumbens/fisiopatología , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/metabolismo , Estrés Psicológico/fisiopatología , Transmisión Sináptica
7.
Glia ; 68(12): 2631-2642, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32585762

RESUMEN

Cortical spreading depression (CSD) is a pathological neural excitation that underlies migraine pathophysiology. Since glutamate receptor antagonists impair CSD propagation, susceptibility to CSD might be determined by any of the neuronal (excitatory amino acid carrier 1 [EAAC1]) and glial (GLutamate ASpartate Transporter [GLAST] and glial glutamate transporter 1 [GLT-1]) glutamate transporters, which are responsible for clearing extracellular glutamate. To investigate this hypothesis, we performed electrophysiological, hemodynamic, and electrochemical analyses using EAAC1- (EAAC1 KO), GLAST- (GLAST KO), and conditional GLT1-1-knockout mice (GLT-1 cKO) to assess altered susceptibility to CSD. Despite the incomplete deletion of the gene in the cerebral cortex, GLT-1 cKO mice exhibited significant reduction of GLT-1 protein in the brain without apparent alteration of the cytoarchitecture in the cerebral cortex. Physiological analysis revealed that GLT-1 cKO showed enhanced susceptibility to CSD elicited by chemical stimulation with increased CSD frequency and velocity compared to GLT-1 control. In contrast, the germ-line EAAC1 and GLAST KOs showed no such effect. Intriguingly, both field potential and cerebral blood flow showed faster dynamics with narrower CSD than the controls. An enzyme-based biosensor revealed more rapid accumulation of glutamate in the extracellular space in GLT-1 cKO mice during the early phase of CSD than in GLT-1 control, resulting in an increased susceptibility to CSD. These results provided the first evidence for a novel role of GLT-1 in determining susceptibility to CSD.


Asunto(s)
Depresión , Animales , Corteza Cerebral/metabolismo , Transportador 2 de Aminoácidos Excitadores , Ácido Glutámico , Ratones
8.
J Neurosci ; 40(28): 5347-5361, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32457076

RESUMEN

Amyloid-ß (Aß) deposition occurs years before cognitive symptoms appear and is considered a cause of Alzheimer's disease (AD). The imbalance of Aß production and clearance leads to Aß accumulation and Aß deposition. Increasing evidence indicates an important role of astrocytes, the most abundant cell type among glial cells in the brain, in Aß clearance. We explored the role of low-density lipoprotein receptor-related protein 4 (LRP4), a member of the LDLR family, in AD pathology. We show that Lrp4 is specifically expressed in astrocytes and its levels in astrocytes were higher than those of Ldlr and Lrp1, both of which have been implicated in Aß uptake. LRP4 was reduced in postmortem brain tissues of AD patients. Genetic deletion of the Lrp4 gene augmented Aß plaques in 5xFAD male mice, an AD mouse model, and exacerbated the deficits in neurotransmission, synchrony between the hippocampus and PFC, and cognition. Mechanistically, LRP4 promotes Aß uptake by astrocytes likely by interacting with ApoE. Together, our study demonstrates that astrocytic LRP4 plays an important role in Aß pathology and cognitive function.SIGNIFICANCE STATEMENT This study investigates how astrocytes, a type of non-nerve cells in the brain, may contribute to Alzheimer's disease (AD) development. We demonstrate that the low-density lipoprotein receptor-related protein 4 (LRP4) is reduced in the brain of AD patients. Mimicking the reduced levels in an AD mouse model exacerbates cognitive impairment and increases amyloid aggregates that are known to damage the brain. We show that LRP4 could promote the clearance of amyloid protein by astrocytes. Our results reveal a previously unappreciated role of LRP4 in AD development.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Enfermedad de Alzheimer/patología , Animales , Astrocitos/patología , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Proteínas Relacionadas con Receptor de LDL/genética , Masculino , Ratones , Placa Amiloide/metabolismo , Placa Amiloide/patología
9.
Neuron ; 105(3): 475-490.e6, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31780330

RESUMEN

Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders with symptoms including social deficits, anxiety, and communication difficulties. However, ASD pathogenic mechanisms are poorly understood. Mutations of CUL3, which encodes Cullin 3 (CUL3), a component of an E3 ligase complex, are thought of as risk factors for ASD and schizophrenia (SCZ). CUL3 is abundant in the brain, yet little is known of its function. Here, we show that CUL3 is critical for neurodevelopment. CUL3-deficient mice exhibited social deficits and anxiety-like behaviors with enhanced glutamatergic transmission and neuronal excitability. Proteomic analysis revealed eIF4G1, a protein for Cap-dependent translation, as a potential target of CUL3. ASD-associated cellular and behavioral deficits could be rescued by pharmacological inhibition of the eIF4G1 function and chemogenetic inhibition of neuronal activity. Thus, CUL3 is critical to neural development, neurotransmission, and excitation-inhibition (E-I) balance. Our study provides novel insight into the pathophysiological mechanisms of ASD and SCZ.


Asunto(s)
Ansiedad/metabolismo , Proteínas Cullin/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Inhibidores/fisiología , Biosíntesis de Proteínas/fisiología , Habilidades Sociales , Animales , Ansiedad/genética , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Proteínas Cullin/genética , Células HEK293 , Humanos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
10.
Elife ; 82019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31268420

RESUMEN

Adult neurogenesis in the hippocampus may represent a form of plasticity in brain functions including mood, learning and memory. However, mechanisms underlying neural stem/progenitor cells (NSPCs) proliferation are not well understood. We found that Agrin, a factor critical for neuromuscular junction formation, is elevated in the hippocampus of mice that are stimulated by enriched environment (EE). Genetic deletion of the Agrn gene in excitatory neurons decreases NSPCs proliferation and increases depressive-like behavior. Low-density lipoprotein receptor-related protein 4 (Lrp4), a receptor for Agrin, is expressed in hippocampal NSPCs and its mutation blocked basal as well as EE-induced NSPCs proliferation and maturation of newborn neurons. Finally, we show that Lrp4 interacts with and activates receptor tyrosine kinase-like orphan receptor 2 (Ror2); and Ror2 mutation impairs NSPCs proliferation. Together, these observations identify a role of Agrin-Lrp4-Ror2 signaling for adult neurogenesis, uncovering previously unexpected functions of Agrin and Lrp4 in the brain.


Asunto(s)
Agrina/metabolismo , Hipocampo/crecimiento & desarrollo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal , Agrina/deficiencia , Animales , Proliferación Celular , Técnicas de Inactivación de Genes , Proteínas Relacionadas con Receptor de LDL/deficiencia , Ratones , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/deficiencia
11.
Proc Natl Acad Sci U S A ; 115(51): 13105-13110, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30498032

RESUMEN

Neurotrophic factor NRG1 and its receptor ErbB4 play a role in GABAergic circuit assembly during development. ErbB4 null mice possess fewer interneurons, have decreased GABA release, and show impaired behavior in various paradigms. In addition, NRG1 and ErbB4 have also been implicated in regulating GABAergic transmission and plasticity in matured brains. However, current ErbB4 mutant strains are unable to determine whether phenotypes in adult mutant mice result from abnormal neural development. This important question, a glaring gap in understanding NRG1-ErbB4 function, was addressed by using two strains of mice with temporal control of ErbB4 deletion and expression, respectively. We found that ErbB4 deletion in adult mice impaired behavior and GABA release but had no effect on neuron numbers and morphology. On the other hand, some deficits due to the ErbB4 null mutation during development were alleviated by restoring ErbB4 expression at the adult stage. Together, our results indicate a critical role of NRG1-ErbB4 signaling in GABAergic transmission and behavior in adulthood and suggest that restoring NRG1-ErbB4 signaling at the postdevelopmental stage might benefit relevant brain disorders.


Asunto(s)
Conducta Animal , Encéfalo/patología , Interneuronas/patología , Neurregulina-1/metabolismo , Receptor ErbB-4/fisiología , Sinapsis/fisiología , Transmisión Sináptica , Animales , Encéfalo/metabolismo , Interneuronas/metabolismo , Ratones , Ratones Noqueados , Neurregulina-1/genética , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo
12.
J Neurosci ; 38(41): 8860-8873, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30171091

RESUMEN

During aging, acetylcholine receptor (AChR) clusters become fragmented and denervated at the neuromuscular junction (NMJ). Underpinning molecular mechanisms are not well understood. We showed that LRP4, a receptor for agrin and critical for NMJ formation and maintenance, was reduced at protein level in aged mice, which was associated with decreased MuSK tyrosine phosphorylation, suggesting compromised agrin-LRP4-MuSK signaling in aged muscles. Transgenic expression of LRP4 in muscles alleviated AChR fragmentation and denervation and improved neuromuscular transmission in aged mice. LRP4 ubiquitination was augmented in aged muscles, suggesting increased LRP4 degradation as a mechanism for reduced LRP4. We found that sarcoglycan α (SGα) interacted with LRP4 and delayed LRP4 degradation in cotransfected cells. AAV9-mediated expression of SGα in muscles mitigated AChR fragmentation and denervation and improved neuromuscular transmission in aged mice. These observations support a model where compromised agrin-LRP4-MuSK signaling serves as a pathological mechanism of age-related NMJ decline and identify a novel function of SGα in stabilizing LRP4 for NMJ stability in aged mice.SIGNIFICANCE STATEMENT This study provides evidence that LRP4, a receptor of agrin that is critical for NMJ formation and maintenance, is reduced at protein level in aged muscles. Transgenic expression of LRP4 in muscles ameliorates AChR fragmentation and denervation and improves neuromuscular transmission in aged mice, demonstrating a critical role of the agrin-LRP4-MuSK signaling. Our study also reveals a novel function of SGα to prevent LRP4 degradation in aged muscles. Finally, we show that NMJ decline in aged mice can be mitigated by AAV9-mediated expression of SGα in muscles. These observations provide insight into pathological mechanisms of age-related NMJ decline and suggest that improved agrin-LRP4-MuSK signaling may be a target for potential therapeutic intervention.


Asunto(s)
Envejecimiento , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , Receptores Colinérgicos/metabolismo , Receptores de LDL/metabolismo , Sarcoglicanos/metabolismo , Animales , Femenino , Proteínas Relacionadas con Receptor de LDL , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/inervación , Fosforilación , Proteínas Tirosina Quinasas Receptoras/metabolismo
13.
Proc Natl Acad Sci U S A ; 115(10): 2508-2513, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463705

RESUMEN

Neuregulin3 (NRG3) is a growth factor of the neuregulin (NRG) family and a risk gene of various severe mental illnesses including schizophrenia, bipolar disorders, and major depression. However, the physiological function of NRG3 remains poorly understood. Here we show that loss of Nrg3 in GFAP-Nrg3f/f mice increased glutamatergic transmission, but had no effect on GABAergic transmission. These phenotypes were observed in Nex-Nrg3f/f mice, where Nrg3 was specifically knocked out in pyramidal neurons, indicating that Nrg3 regulates glutamatergic transmission by a cell-autonomous mechanism. Consequently, in the absence of Nrg3 in pyramidal neurons, mutant mice displayed various behavioral deficits related to mental illnesses. We show that the Nrg3 mutation decreased paired-pulse facilitation, increased decay of NMDAR currents when treated with MK801, and increased minimal stimulation-elicited response, providing evidence that the Nrg3 mutation increases glutamate release probability. Notably, Nrg3 is a presynaptic protein that regulates the SNARE-complex assembly. Finally, increased Nrg3 levels, as observed in patients with severe mental illnesses, suppressed glutamatergic transmission. Together, these observations indicate that, unlike the prototype Nrg1, the effect of which is mediated by activating ErbB4 in interneurons, Nrg3 is critical in controlling glutamatergic transmission by regulating the SNARE complex at the presynaptic terminals, identifying a function of Nrg3 and revealing a pathophysiological mechanism for hypofunction of the glutamatergic pathway in Nrg3-related severe mental illnesses.


Asunto(s)
Ácido Glutámico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas SNARE/metabolismo , Animales , Conducta Animal/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Trastornos Mentales/genética , Ratones , Ratones Transgénicos , Neurregulinas , Células Piramidales/metabolismo
14.
Hum Mol Genet ; 25(20): 4507-4517, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28173004

RESUMEN

Point mutations in the vacuolar protein sorting 35 gene (VPS35) have been associated with an autosomal dominant form of late-onset Parkinson disease (PARK17), but there has been considerable debate over whether it is caused by a loss- or gain-of-function mechanism and over the intracellular target site of neurotoxicity. To investigate the pathogenesis of PARK17 in vivo, we generated Vps35 D620N knock-in (KI) mice, expressing the homologous mutant protein with endogenous patterns of expression, simultaneously with Vps35 deletion 1 (Del1) mice, which carry 1bp deletion in the exon15 of Vps35, by CRISPR/Cas9-mediated genome engineering. Neither homozygous nor heterozygous Vps35 D620N KI mice suffered from premature death or developed clear neurodegeneration up to 70 weeks of age. Vps35 Del1 allele appeared to be a null or at least severely hypomorphic allele and homozygous Vps35 Del1 showed early embryonic lethality. Heterozygous crossings between Del1 and D620N knock-in mice revealed that the D620N/Del1 compound heterozygous mice, but not heterozygous Del1 mice, suffered from survival disadvantage. In vivo microdialysis showed that DA release evoked by 120 mM potassium chloride was significantly reduced in the caudate putamen of adult homozygous Vps35 D620N KI mice. Taken together, these results suggest that Vps35 D620N allele is a partial-loss-of-function allele and that such a genetic predisposition and age-related alterations in the nigrostriatal dopamine system cooperatively influence the pathogenesis of PARK17.


Asunto(s)
Modelos Animales de Enfermedad , Dopamina/metabolismo , Mutación , Enfermedad de Parkinson/metabolismo , Proteínas de Transporte Vesicular/genética , Animales , Técnicas de Sustitución del Gen , Homocigoto , Ratones , Neostriado/metabolismo , Neostriado/fisiopatología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/fisiopatología
15.
Neuropsychopharmacology ; 40(7): 1569-79, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25662838

RESUMEN

An increase in the ratio of cellular excitation to inhibition (E/I ratio) has been proposed to underlie the pathogenesis of neuropsychiatric disorders, such as autism spectrum disorders (ASD), obsessive-compulsive disorder (OCD), and Tourette's syndrome (TS). A proper E/I ratio is achieved via factors expressed in neuron and glia. In astrocytes, the glutamate transporter GLT1 is critical for regulating an E/I ratio. However, the role of GLT1 dysfunction in the pathogenesis of neuropsychiatric disorders remains unknown because mice with a complete deficiency of GLT1 exhibited seizures and premature death. Here, we show that astrocyte-specific GLT1 inducible knockout (GLAST(CreERT2/+)/GLT1(flox/flox), iKO) mice exhibit pathological repetitive behaviors including excessive and injurious levels of self-grooming and tic-like head shakes. Electrophysiological studies reveal that excitatory transmission at corticostriatal synapse is normal in a basal state but is increased after repetitive stimulation. Furthermore, treatment with an N-methyl-D-aspartate (NMDA) receptor antagonist memantine ameliorated the pathological repetitive behaviors in iKO mice. These results suggest that astroglial GLT1 has a critical role in controlling the synaptic efficacy at corticostriatal synapses and its dysfunction causes pathological repetitive behaviors.


Asunto(s)
Corteza Cerebral/patología , Trastornos de Traumas Acumulados/genética , Trastornos de Traumas Acumulados/patología , Transportador 1 de Aminoácidos Excitadores/deficiencia , Transportador 2 de Aminoácidos Excitadores/deficiencia , Sinapsis/genética , Animales , Animales Recién Nacidos , Ansiedad/genética , Trastornos de Traumas Acumulados/complicaciones , Trastornos de Traumas Acumulados/tratamiento farmacológico , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/genética , Potenciales Postsinápticos Excitadores/genética , Femenino , Regulación de la Expresión Génica/genética , Hiperalgesia/genética , Masculino , Ratones , Ratones Transgénicos , Degeneración Nerviosa/etiología , Degeneración Nerviosa/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas/genética , Convulsiones/genética
16.
J Neurosci ; 34(49): 16273-85, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471567

RESUMEN

The lateral habenula (LHb) regulates the activity of monoaminergic neurons in the brainstem. This area has recently attracted a surge of interest in psychiatry because studies have reported the pathological activation of the habenula in patients with major depression and in animal models. The LHb plays a significant role in the pathophysiology of depression; however, how habenular neurons are activated to cause various depression symptoms, such as reduced motivation and sleep disturbance, remain unclear. We hypothesized that dysfunctional astrocytes may cause LHb hyperactivity due to the defective uptake activity of extracellular glutamate, which induces depressive-like behaviors. We examined the activity of neurons in habenular pathways and performed behavioral and sleep analyses in mice with pharmacological and genetic inhibition of the activity of the glial glutamate transporter GLT-1 in the LHb. The habenula-specific inhibition of GLT-1 increased the neuronal firing rate and the level of c-Fos expression in the LHb. Mice with reduced GLT-1 activity in the habenula exhibited a depressive-like phenotype in the tail suspension and novelty-suppressed feeding tests. These animals also displayed increased susceptibility to chronic stress, displaying more frequent avoidant behavior without affecting locomotor activity in the open-field test. Intriguingly, the mice showed disinhibition of rapid eye movement sleep, which is a characteristic sleep pattern in patients with depression. These results provide evidence that disrupting glutamate clearance in habenular astrocytes increases neuronal excitability and depressive-like phenotypes in behaviors and sleep.


Asunto(s)
Astrocitos/patología , Depresión/patología , Depresión/fisiopatología , Habénula/fisiopatología , Sueño REM , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Depresión/genética , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 2 de Aminoácidos Excitadores/genética , Habénula/efectos de los fármacos , Habénula/patología , Ratones , Ratones Transgénicos , Microinyecciones , Neuronas/fisiología , Inhibidores de la Captación de Neurotransmisores/administración & dosificación , Inhibidores de la Captación de Neurotransmisores/farmacología , Pironas/administración & dosificación , Pironas/farmacología , Sueño REM/genética
17.
Front Hum Neurosci ; 7: 826, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24339810

RESUMEN

Depression occurs frequently with sleep disturbance such as insomnia. Sleep in depression is associated with disinhibition of the rapid eye movement (REM) sleep. Despite the coincidence of the depression and sleep disturbance, neural substrate for depressive behaviors and sleep regulation remains unknown. Habenula is an epithalamic structure regulating the activities of monoaminergic neurons in the brain stem. Since the imaging studies showed blood flow increase in the habenula of depressive patients, hyperactivation of the habenula has been implicated in the pathophysiology of the depression. Recent electrophysiological studies reported a novel role of the habenular structure in regulation of REM sleep. In this article, we propose possible cellular mechanisms which could elicit the hyperactivation of the habenular neurons and a hypothesis that dysfunction in the habenular circuit causes the behavioral and sleep disturbance in depression. Analysis of the animals with hyperactivated habenula would open the door to understand roles of the habenula in the heterogeneous symptoms such as reduced motor behavior and altered REM sleep in depression.

18.
Respir Physiol Neurobiol ; 175(1): 97-103, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-20870044

RESUMEN

BALB/c mice were sensitized and challenged with ovalbumin. We hypothesized that Kidins220/ARMS influences airway inflammation and hyper-responsiveness during allergic airway challenge, and assessed it by intranasal administration of anti-NGF antibody or anti-ARMS antibody to mice. Airway resistance was measured using a sealed whole-body plethysmograph. Total cell numbers and the percentage of different inflammatory cells in BALF were counted. Expression of IL-1ß, IL-4 and TNF-α were determined by ELISA, and NF-κB activation determined by EMSA. Kidins220/ARMS expression was observed in ovalbumin-sensitized mice by immunofluorescence or western blotting. IL-1ß, IL-4, and TNF-α were overexpressed and NF-κB activation increased after allergen challenge compared with controls. After treatment with anti-ARMS or anti-NGF, levels of IL-1ß, IL-4 and TNF-α and NF-κB activation were reduced in comparison with those of ovalbumin-sensitized mice. These results suggest that NGF-mediated Kidins220/ARMS signaling participates in the pathogenesis of asthma, and contributes to airway inflammation and hyper-responsiveness in ovalbumin-sensitized mice.


Asunto(s)
Proteínas de la Membrana/metabolismo , Ovalbúmina/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Animales , Anticuerpos/administración & dosificación , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática/métodos , Inflamación/inmunología , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Factor de Crecimiento Nervioso/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
19.
Ann Allergy Asthma Immunol ; 105(4): 299-306, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20934630

RESUMEN

BACKGROUND: Nerve growth factor (NGF), combined with its high-affinity receptor tyrosine kinase receptor A (TrkA), has been reported to be involved in the pathogenesis of asthma. OBJECTIVE: To investigate whether the downstream protein ankyrin-rich membrane spanning (ARMS), a novel transmembrane substrate of protein kinase D (Kidins220), is activated in the pathogenesis of asthma. METHODS: The asthmatic model was established by the inhalation of ovalbumin in BALB/c mice. The effects of NGF and TrkA on Kidins220/ARMS in an allergic airway challenge were assessed by administering anti-NGF or anti-TrkA antibody to the mice. Pathologic changes in the bronchi and lung tissues were examined by means of hematoxylin and eosin staining; the inflammatory cells in the bronchoalveolar lavage fluid (BALF) were counted; and co-expression of ARMS and TrkA in BALF cells was observed by means of immunofluorescence. In addition, Kidins220/ARMS, CrkL, NGF, TrkA protein, and Kidins220 messenger RNA levels were determined using Western blot or quantitative reverse transcription-polymerase chain reaction. RESULTS: Using fluorescence microscopy, we found that Kidins220 and TrkA were co-expressed on the membranes of the BALF cells of asthmatic mice. Compared with expression in control animals, Kidins220/ARMS, CrkL, NGF, and TrkA were overexpressed in the lungs after allergen challenge. Moreover, after the mice were treated with anti-NGF or anti-TrkA, the Kidins220/ARMS levels and allergen-induced airway inflammation decreased. CONCLUSIONS: These results suggest that Kidins220/ARMS partly participates in the pathogenesis of asthma through the NGF-TrkA signaling pathway, possibly representing a new mechanism in asthma.


Asunto(s)
Ancirinas/metabolismo , Asma/inmunología , Bronquios/metabolismo , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Animales , Ancirinas/genética , Ancirinas/inmunología , Anticuerpos Monoclonales/administración & dosificación , Asma/metabolismo , Asma/terapia , Bronquios/efectos de los fármacos , Bronquios/inmunología , Bronquios/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/inmunología , Receptor trkA/genética , Receptor trkA/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA