Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 163(5): 1153-1164, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26590419

RESUMEN

Fear is induced by innate and learned mechanisms involving separate pathways. Here, we used an olfactory-mediated innate-fear versus learned-fear paradigm to investigate how these pathways are integrated. Notably, prior presentation of innate-fear stimuli inhibited learned-freezing response, but not vice versa. Whole-brain mapping and pharmacological screening indicated that serotonin-2A receptor (Htr2a)-expressing cells in the central amygdala (CeA) control both innate and learned freezing, but in opposing directions. In vivo fiber photometry analyses in freely moving mice indicated that innate but not learned-fear stimuli suppressed the activity of Htr2a-expressing CeA cells. Artificial inactivation of these cells upregulated innate-freezing response and downregulated learned-freezing response. Thus, Htr2a-expressing CeA cells serve as a hierarchy generator, prioritizing innate fear over learned fear.


Asunto(s)
Amígdala del Cerebelo/fisiología , Miedo/fisiología , Vías Nerviosas , Receptor de Serotonina 5-HT2A/metabolismo , Animales , Miedo/clasificación , Integrasas , Masculino , Ratones , Ratones Endogámicos C57BL , Sustancia Gris Periacueductal/fisiología , Receptor de Serotonina 5-HT2A/genética , Olfato
2.
Commun Biol ; 4(1): 101, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33483561

RESUMEN

Innate fear intimately connects to the life preservation in crises, although this relationships is not fully understood. Here, we report that presentation of a supernormal innate fear inducer 2-methyl-2-thiazoline (2MT), but not learned fear stimuli, induced robust systemic hypothermia/hypometabolism and suppressed aerobic metabolism via phosphorylation of pyruvate dehydrogenase, thereby enabling long-term survival in a lethal hypoxic environment. These responses exerted potent therapeutic effects in cutaneous and cerebral ischemia/reperfusion injury models. In contrast to hibernation, 2MT stimulation accelerated glucose uptake in the brain and suppressed oxygen saturation in the blood. Whole-brain mapping and chemogenetic activation revealed that the sensory representation of 2MT orchestrates physiological responses via brain stem Sp5/NST to midbrain PBN pathway. 2MT, as a supernormal stimulus of innate fear, induced exaggerated, latent life-protective effects in mice. If this system is preserved in humans, it may be utilized to give rise to a new field: "sensory medicine."


Asunto(s)
Miedo/fisiología , Hibernación , Instinto , Odorantes , Tiazoles , Animales , Evaluación Preclínica de Medicamentos , Hipotermia Inducida , Hipoxia , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión/prevención & control
3.
Nat Commun ; 12(1): 2074, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824316

RESUMEN

Thiazoline-related innate fear-eliciting compounds (tFOs) orchestrate hypothermia, hypometabolism, and anti-hypoxia, which enable survival in lethal hypoxic conditions. Here, we show that most of these effects are severely attenuated in transient receptor potential ankyrin 1 (Trpa1) knockout mice. TFO-induced hypothermia involves the Trpa1-mediated trigeminal/vagal pathways and non-Trpa1 olfactory pathway. TFOs activate Trpa1-positive sensory pathways projecting from trigeminal and vagal ganglia to the spinal trigeminal nucleus (Sp5) and nucleus of the solitary tract (NTS), and their artificial activation induces hypothermia. TFO presentation activates the NTS-Parabrachial nucleus pathway to induce hypothermia and hypometabolism; this activation was suppressed in Trpa1 knockout mice. TRPA1 activation is insufficient to trigger tFO-mediated anti-hypoxic effects; Sp5/NTS activation is also necessary. Accordingly, we find a novel molecule that enables mice to survive in a lethal hypoxic condition ten times longer than known tFOs. Combinations of appropriate tFOs and TRPA1 command intrinsic physiological responses relevant to survival fate.


Asunto(s)
Miedo/fisiología , Hipotermia/metabolismo , Hipoxia/metabolismo , Canal Catiónico TRPA1/metabolismo , Tiazoles/farmacología , Animales , Bradicardia/patología , Miedo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Hipotermia/complicaciones , Hipoxia/complicaciones , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Sensación/efectos de los fármacos , Tiazoles/química , Factores de Tiempo , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo , Nervio Vago/efectos de los fármacos
4.
Psychiatry Res ; 168(2): 119-28, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19501919

RESUMEN

Fyn, a Src-family kinase, is highly expressed in brain tissue and blood cells. In the mouse brain, Fyn participates in brain development, synaptic transmission through the phosphorylation of N-methyl-d-aspartate (NMDA) receptor subunits, and the regulation of emotional behavior. Recently, we found that Fyn is required for the signal transduction in striatal neurons that is initiated by haloperidol, an antipsychotic drug. To determine whether Fyn abnormalities are present in patients with schizophrenia, we analyzed Fyn expression in platelet samples from 110 patients with schizophrenia, 75 of the patients' first-degree relatives, and 130 control subjects. A Western blot analysis revealed significantly lower levels of Fyn protein among the patients with schizophrenia and their relatives, compared with the level in the control group. At the mRNA level, the splicing patterns of fyn were altered in the patients and their relatives; specifically, the ratio of fynDelta7, in which exon 7 is absent, was elevated. An expression study in HEK293T cells revealed that FynDelta7 had a dominant-negative effect on the phosphorylation of Fyn's substrate. These results suggest novel deficits in Fyn function, manifested as the downregulation of Fyn protein or the altered transcription of the fyn gene, in patients with schizophrenia.


Asunto(s)
Plaquetas/metabolismo , Expresión Génica , Proteínas Proto-Oncogénicas c-fyn/genética , Esquizofrenia/genética , Adulto , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Animales , Western Blotting , Regulación hacia Abajo/genética , Exonucleasas/genética , Exonucleasas/metabolismo , Familia , Femenino , Humanos , Inteínas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esquizofrenia/sangre , Activación Transcripcional/genética
5.
Eur J Neurosci ; 28(5): 973-81, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18691323

RESUMEN

Fyn-tyrosine-kinase-deficient mice exhibit defects in the Morris water maze test and long-term potentiation (LTP) induction in the hippocampus, and given that LTP has been postulated as the neural basis for memory formation, Fyn may be required for hippocampus-dependent memory formation. However, how Fyn is involved in the process of memory formation is unclear. To investigate the role of Fyn in hippocampal memory formation, we first tested the behavior of Fyn-deficient mice by contextual fear conditioning. A mouse was placed in a context and a foot shock was delivered, so that the mouse associated the context with the shock. We found that the freezing response of Fyn-deficient mice to the context was impaired at 24 h after conditioning. We then measured freezing at 1 h after conditioning, and found that their short-term contextual fear memory was also impaired. We used Western blotting to examine the mode of Fyn activation in dorsal hippocampal tissue following contextual fear conditioning. Fyn activation peaked as early as 5-10 min after contextual fear conditioning and persisted for at least 40 min. Concomitant increases in tyrosine phosphorylation of several proteins, including NR2B, were also observed, but no increases in tyrosine phosphorylation were observed in Fyn-deficient mice. Thus, both short-term and long-term (24-h) contextual fear memory were impaired in Fyn-deficient mice, and Fyn activation in the dorsal hippocampus transiently increased after contextual fear conditioning. These findings strongly suggest that activation of the Fyn signaling pathway is involved in hippocampus-dependent formation of contextual fear memory.


Asunto(s)
Reacción de Prevención/fisiología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Hipocampo/enzimología , Memoria/fisiología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Animales , Estimulación Eléctrica , Activación Enzimática/fisiología , Hipocampo/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
6.
Brain Res ; 1073-1074: 11-9, 2006 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-16438943

RESUMEN

Reelin is a large glycoprotein that is secreted into the extracellular matrix. In the embryonic brain, the binding of Reelin to its receptors ApoER2 and VLDLR induces subcellular events that include the activation Fyn tyrosine kinase, and plays a crucial role in cortical formation. Reelin signaling is also involved in postnatal brain functions such as dendrite development and synaptic plasticity. However, the molecular events involved in Reelin signaling in the postnatal brain remain to be elucidated. Here, we evaluated the proteins downstream of Reelin signaling by comparing the tyrosine-phosphorylated proteins in the postnatal hippocampus of heterozygous and homozygous reeler and wild-type mice, by Western blot analyses. We found that the levels of several phosphoproteins were highest in the hippocampus of the heterozygous reeler mice. The most prominent increase was of two 180-kDa phosphoproteins, which were identified as the NR2A and NR2B subunits of NMDA-R. The amounts of these proteins also increased in the hippocampus of heterozygous reeler mice. However, the mRNA levels of the NMDA-R subunits, determined by quantitative RT-PCR, were the same as in wild-type mice. We also found that the increase in NR2A and NR2B proteins in heterozygous reeler was dependent on Fyn, because this change was absent in heterozygous reeler/homozygous Fyn-deficient double-mutant mice. Thus, the NMDA-R protein level is regulated by the Reelin protein level in a Fyn-dependent manner in the mouse brain.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Heterocigoto , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Embrión de Mamíferos , Inmunoprecipitación/métodos , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/deficiencia , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/clasificación , Receptores de N-Metil-D-Aspartato/genética , Proteína Reelina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
7.
Gene ; 349: 1-14, 2005 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-15777644

RESUMEN

The human protocadherin (Pcdh) gene clusters are located on chromosome 5q31. Single-nucleotide polymorphisms (SNPs) were detected in the Pcdh-alpha and -beta variable exons, and in the Pcdh-alpha constant exon, in samples from 104 individuals. Among coding SNPs (cSNPs), nonsynonymous (amino acid exchange) SNPs were 2.2 times more common than synonymous (silent) changes in the Pcdh-alpha variable exons, but only 1.2 times more common in the Pcdh-beta variable exons. The nonsynonymous SNPs were high in the ectodomain (EC) 1 encoding region of Pcdh-alpha but not of Pcdh-beta. One 48-kb region of extensive linkage disequilibrium (LD) is reported that has two haplotypes extending from the alpha1 to alpha7 genes in the Pcdh-alpha cluster. Here we identified 15 amino acid exchanges in these two major haplotypes; therefore, the two haplotypes encode different sets of Pcdh-alpha proteins in the brain. The distribution of cSNPs was different for each EC region of Pcdh-alpha or -beta. The frequency of cSNPs was negatively correlated with the paralogous sequence diversity. These results suggested that gene conversion events in homologous regions of the Pcdh-alpha and Pcdh-beta clusters generated the cSNPs. Within the cSNPs, gene conversions were found in Pcdh-alpha4 in the major haplotype, and in Pcdh-beta9. These gene conversions were caused by the unequal crossing-over of homologous sequence regions. Thus, nonsynonymous variations in the Pcdh-alpha and -beta genes are possible contributors to the variations in human brain function.


Asunto(s)
Cadherinas/genética , Familia de Multigenes , Polimorfismo de Nucleótido Simple , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Pueblo Asiatico , Encéfalo/fisiología , Cadherinas/química , Mapeo Cromosómico , Cromosomas Humanos Par 5 , Secuencia Conservada , ADN/sangre , ADN/genética , Evolución Molecular , Exones , Conversión Génica , Frecuencia de los Genes , Variación Genética , Haplotipos , Humanos , Desequilibrio de Ligamiento , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN , Eliminación de Secuencia , Homología de Secuencia de Aminoácido
8.
Neuroreport ; 21(8): 554-8, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20400923

RESUMEN

Contextual fear memory is attenuated by re-exposure of animals to a context alone without pairing it with an unconditioned stimulus, and this phenomenon is referred to as fear extinction. In this study we demonstrated that stereotaxic injection of an inhibitor of Src homology 2-containing protein-tyrosine phosphatases 1 and 2 (SHP1/2), NSC87877, into the hippocampus significantly suppressed extinction of contextual fear in the mouse. Intra-hippocampal injection of NSC87877, however, had no effect on the initial memory formation in contextual fear conditioning. These findings suggest that SHP1/2 activity in the hippocampus is involved in the control of contextual fear extinction.


Asunto(s)
Extinción Psicológica/fisiología , Miedo/fisiología , Hipocampo/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Quinolinas/farmacología
9.
Neuroreport ; 20(16): 1461-5, 2009 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-19752763

RESUMEN

Contextual fear memory is attenuated by reexposure of animals to a context alone without pairing it with an unconditioned stimulus, a phenomenon referred to as fear extinction. Here, we report that Fyn tyrosine kinase in the hippocampus is involved in the extinction of contextual fear. We inhibited Src-family tyrosine kinases in the dorsal hippocampus by stereotaxic injection of an inhibitor, PP2, and observed facilitation of extinction. We then biochemically analyzed dorsal hippocampal tissue during extinction training, and found that activated Fyn was significantly downregulated among the Src-family tyrosine kinases examined. These findings suggest that downregulation of Fyn activity facilitates the extinction of contextual fear.


Asunto(s)
Condicionamiento Clásico/fisiología , Extinción Psicológica/fisiología , Miedo , Hipocampo/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Análisis de Varianza , Animales , Conducta Animal , Condicionamiento Clásico/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Extinción Psicológica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Factores de Tiempo
10.
J Biol Chem ; 281(11): 7129-35, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16407246

RESUMEN

Fyn-mediated tyrosine phosphorylation of N-methyl-D-aspartate (NMDA) receptor subunits has been implicated in various brain functions, including ethanol tolerance, learning, and seizure susceptibility. In this study, we explored the role of Fyn in haloperidol-induced catalepsy, an animal model of the extrapyramidal side effects of antipsychotics. Haloperidol induced catalepsy and muscle rigidity in the control mice, but these responses were significantly reduced in Fyn-deficient mice. Expression of the striatal dopamine D(2) receptor, the main site of haloperidol action, did not differ between the two genotypes. Fyn activation and enhanced tyrosine phosphorylation of the NMDA receptor NR2B subunit, as measured by Western blotting, were induced after haloperidol injection of the control mice, but both responses were significantly reduced in Fyn-deficient mice. Dopamine D(2) receptor blockade was shown to increase both NR2B phosphorylation and the NMDA-induced calcium responses in control cultured striatal neurons but not in Fyn-deficient neurons. Based on these findings, we proposed a new molecular mechanism underlying haloperidol-induced catalepsy, in which the dopamine D(2) receptor antagonist induces striatal Fyn activation and the subsequent tyrosine phosphorylation of NR2B alters striatal neuronal activity, thereby inducing the behavioral changes that are manifested as a cataleptic response.


Asunto(s)
Catalepsia/inducido químicamente , Haloperidol/farmacología , Proteínas Proto-Oncogénicas c-fyn/fisiología , Animales , Antidiscinéticos/farmacología , Antipsicóticos/farmacología , Western Blotting , Calcio/metabolismo , Catalepsia/patología , Células Cultivadas , Dopamina/metabolismo , Genotipo , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Indoles/farmacología , Ratones , Neuronas/metabolismo , Fosforilación , Piperidinas/farmacología , Proteínas Tirosina Quinasas/química , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de N-Metil-D-Aspartato/química , Transducción de Señal , Factores de Tiempo , Tirosina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA