Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Neuron ; 112(4): 611-627.e8, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38086372

RESUMO

Social animals compete for limited resources, resulting in a social hierarchy. Although different neuronal subpopulations in the medial prefrontal cortex (mPFC), which has been mechanistically implicated in social dominance behavior, encode distinct social competition behaviors, their identities and associated molecular underpinnings have not yet been identified. In this study, we found that mPFC neurons projecting to the nucleus accumbens (mPFC-NAc) encode social winning behavior, whereas mPFC neurons projecting to the ventral tegmental area (mPFC-VTA) encode social losing behavior. High-throughput single-cell transcriptomic analysis and projection-specific genetic manipulation revealed that the expression level of POU domain, class 3, transcription factor 1 (Pou3f1) in mPFC-VTA neurons controls social hierarchy. Optogenetic activation of mPFC-VTA neurons increases Pou3f1 expression and lowers social rank. Together, these data demonstrate that discrete activity and gene expression in separate mPFC projections oppositely orchestrate social competition and hierarchy.


Assuntos
Núcleo Accumbens , Área Tegmentar Ventral , Animais , Área Tegmentar Ventral/fisiologia , Núcleo Accumbens/fisiologia , Comportamento Social , Córtex Pré-Frontal/fisiologia , Neurônios
2.
Exp Neurobiol ; 32(5): 313-327, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37927130

RESUMO

Mental health is influenced by the gut-brain axis; for example, gut dysbiosis has been observed in patients with major depressive disorder (MDD). Gut microbial changes by fecal microbiota transplantation or probiotics treatment reportedly modulates depressive symptoms. However, it remains unclear how gut dysbiosis contributes to mental dysfunction, and how correction of the gut microbiota alleviates neuropsychiatric disorders. Our previous study showed that chronic consumption of Lactobacillus reuteri ATG-F4 (F4) induced neurometabolic alterations in healthy mice. Here, we investigated whether F4 exerted therapeutic effects on depressive-like behavior by influencing the central nervous system. Using chronic unpredictable stress (CUS) to induce anhedonia, a key symptom of MDD, we found that chronic F4 consumption alleviated CUS-induced anhedonic behaviors, accompanied by biochemical changes in the gut, serum, and brain. Serum and brain metabolite concentrations involved in tryptophan metabolism were regulated by CUS and F4. F4 consumption reduced the elevated levels of serotonin (5-HT) in the brain observed in the CUS group. Additionally, the increased expression of Htr1a, a subtype of the 5-HT receptor, in the medial prefrontal cortex (mPFC) of stressed mice was restored to levels observed in stress-naïve mice following F4 supplementation. We further demonstrated the role of Htr1a using AAV-shRNA to downregulate Htr1a in the mPFC of CUS mice, effectively reversing CUS-induced anhedonic behavior. Together, our findings suggest F4 as a potential therapeutic approach for relieving some depressive symptoms and highlight the involvement of the tryptophan metabolism in mitigating CUS-induced depressive-like behaviors through the action of this bacterium.

3.
Biol Psychiatry ; 92(2): 104-115, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35314057

RESUMO

BACKGROUND: Metabotropic glutamate receptor 5 (mGluR5) has been implicated in stress-related psychiatric disorders, particularly major depressive disorder. Although growing evidence supports the proresilient role of mGluR5 in corticolimbic circuitry in the depressive-like behaviors following chronic stress exposure, the underlying neural mechanisms, including circuits and molecules, remain unknown. METHODS: We measured the c-Fos expression and probability of neurotransmitter release in and from basolateral amygdala (BLA) neurons projecting to the medial prefrontal cortex (mPFC) and to the ventral hippocampus (vHPC) after chronic social defeat stress. The role of BLA projections in depressive-like behaviors was assessed using optogenetic manipulations, and the underlying molecular mechanisms of mGluR5 and downstream signaling were investigated by Western blotting, viral-mediated gene transfer, and pharmacological manipulations. RESULTS: Chronic social defeat stress disrupted neural activity and glutamatergic transmission in both BLA projections. Optogenetic activation of BLA projections reversed the detrimental effects of chronic social defeat stress on depressive-like behaviors and mGluR5 expression in the mPFC and vHPC. Conversely, inhibition of BLA projections of mice undergoing subthreshold social defeat stress induced a susceptible phenotype and mGluR5 reduction. These two BLA circuits appeared to act in an independent way. We demonstrate that mGluR5 overexpression in the mPFC or vHPC was proresilient while the mGluR5 knockdown was prosusceptible and that the proresilient effects of mGluR5 are mediated through distinctive downstream signaling pathways in the mPFC and vHPC. CONCLUSIONS: These findings identify mGluR5 in the mPFC and vHPC that receive BLA inputs as a critical mediator of stress resilience, highlighting circuit-specific signaling for depressive-like behaviors.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Receptor de Glutamato Metabotrópico 5 , Estresse Psicológico , Tonsila do Cerebelo/metabolismo , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Transtorno Depressivo Maior/metabolismo , Humanos , Camundongos , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo
4.
Commun Biol ; 4(1): 1138, 2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34588597

RESUMO

Many synaptic adhesion molecules positively regulate synapse development and function, but relatively little is known about negative regulation. SALM4/Lrfn3 (synaptic adhesion-like molecule 4/leucine rich repeat and fibronectin type III domain containing 3) inhibits synapse development by suppressing other SALM family proteins, but whether SALM4 also inhibits synaptic function and specific behaviors remains unclear. Here we show that SALM4-knockout (Lrfn3-/-) male mice display enhanced contextual fear memory consolidation (7-day post-training) but not acquisition or 1-day retention, and exhibit normal cued fear, spatial, and object-recognition memory. The Lrfn3-/- hippocampus show increased currents of GluN2B-containing N-methyl-D-aspartate (NMDA) receptors (GluN2B-NMDARs), but not α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors (AMPARs), which requires the presynaptic receptor tyrosine phosphatase PTPσ. Chronic treatment of Lrfn3-/- mice with fluoxetine, a selective serotonin reuptake inhibitor used to treat excessive fear memory that directly inhibits GluN2B-NMDARs, normalizes NMDAR function and contextual fear memory consolidation in Lrfn3-/- mice, although the GluN2B-specific NMDAR antagonist ifenprodil was not sufficient to reverse the enhanced fear memory consolidation. These results suggest that SALM4 suppresses excessive GluN2B-NMDAR (not AMPAR) function and fear memory consolidation (not acquisition).


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Medo/fisiologia , Consolidação da Memória/fisiologia , Receptores de N-Metil-D-Aspartato/genética , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Camundongos , Camundongos Knockout , Receptores de N-Metil-D-Aspartato/metabolismo
5.
Exp Neurobiol ; 29(6): 403-416, 2020 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-33139585

RESUMO

Often called the second brain, the gut communicates extensively with the brain and vice versa. The conversation between these two organs affects a variety of physiological mechanisms that are associated with our mental health. Over the past decade, a growing body of evidence has suggested that the gut microbiome builds a unique ecosystem inside the gastrointestinal tract to maintain the homeostasis and that compositional changes in the gut microbiome are highly correlated with several mental disorders. There are ongoing efforts to treat or prevent mental disorders by regulating the gut microbiome using probiotics. These attempts are based on the seminal findings that probiotics can control the gut microbiome and affect mental conditions. However, some issues have yet to be conclusively addressed, especially the causality between the gut microbiome and mental disorders. In this review, we focus on the mechanisms by which the gut microbiome affects mental health and diseases. Furthermore, we discuss the potential use of probiotics as therapeutic agents for psychiatric disorders.

6.
Cell Rep ; 28(5): 1154-1166.e5, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31365861

RESUMO

The underlying mechanisms of how positive emotional valence (e.g., pleasure) causes preference of an associated context is poorly understood. Here, we show that activation of astrocytic µ-opioid receptor (MOR) drives conditioned place preference (CPP) by means of specific modulation of astrocytic MOR, an exemplar endogenous Gi protein-coupled receptor (Gi-GPCR), in the CA1 hippocampus. Long-term potentiation (LTP) induced by a subthreshold stimulation with the activation of astrocytic MOR at the Schaffer collateral pathway accounts for the memory acquisition to induce CPP. This astrocytic MOR-mediated LTP induction is dependent on astrocytic glutamate released upon activation of the astrocytic MOR and the consequent activation of the presynaptic mGluR1. The astrocytic MOR-dependent LTP and CPP were recapitulated by a chemogenetic activation of astrocyte-specifically expressed Gi-DREADD hM4Di. Our study reveals that the transduction of inhibitory Gi-signaling into augmented excitatory synaptic transmission through astrocytic glutamate is critical for the acquisition of contextual memory for CPP.


Assuntos
Astrócitos/metabolismo , Região CA1 Hipocampal/metabolismo , Memória , Receptores Opioides mu/metabolismo , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Knockout , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Opioides mu/genética
7.
Mol Psychiatry ; 24(8): 1189-1205, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30651602

RESUMO

Recent genome-wide association studies on major depressive disorder have implicated neuronal growth regulator 1 (Negr1), a GPI-anchored cell adhesion molecule in the immunoglobulin LON family. Although Negr1 has been shown to regulate neurite outgrowth and synapse formation, the mechanism through which this protein affects mood disorders is still largely unknown. In this research, we characterized Negr1-deficient (negr1-/-) mice to elucidate the function of Negr1 in anxiety and depression. We found that anxiety- and depression-like behaviors increased in negr1-/- mice compared with wild-type mice. In addition, negr1-/- mice had decreased adult hippocampal neurogenesis compared to wild-type mice. Concurrently, both LTP and mEPSC in the dentate gyrus (DG) region were severely compromised in negr1-/- mice. In our effort to elucidate the underlying molecular mechanisms, we found that lipocalin-2 (Lcn2) expression was decreased in the hippocampus of negr1-/- mice compared to wild-type mice. Heterologous Lcn2 expression in the hippocampal DG of negr1-/- mice rescued anxiety- and depression-like behaviors and restored neurogenesis and mEPSC frequency to their normal levels in these mice. Furthermore, we discovered that Negr1 interacts with leukemia inhibitory factor receptor (LIFR) and modulates LIF-induced Lcn2 expression. Taken together, our data uncovered a novel mechanism of mood regulation by Negr1 involving an interaction between Negr1 and LIFR along with Lcn2 expression.


Assuntos
Ansiedade/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Depressão/genética , Animais , Ansiedade/fisiopatologia , Transtornos de Ansiedade/genética , Transtornos de Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Giro Denteado/metabolismo , Depressão/fisiopatologia , Transtorno Depressivo/genética , Transtorno Depressivo/fisiopatologia , Estudo de Associação Genômica Ampla , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/fisiologia , Lobo Temporal/metabolismo
8.
Sci Rep ; 8(1): 17653, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30518785

RESUMO

Thalidomide is a widely prescribed immunomodulatory drug (iMiD) for multiple myeloma, but causes reversible memory loss in humans. However, how thalidomide causes cognitive dysfunction at a cellular and molecular level has not been demonstrated. We studied the effect of thalidomide on synaptic functions and cognitive behaviors using a mouse model. Thalidomide led to cognitive deficits in learning behavior in a passive avoidance test and in a novel object recognition test, increased anxiety in an elevated plus maze test, and increased depressive behaviors in a tail suspension test. Interestingly, thalidomide elevated big- or large-conductance, calcium-activated K+ (BK) channel expression in the plasma membrane and BK channel activity in the hippocampus. Thalidomide also increased the paired pulse ratio of excitatory postsynaptic current (EPSC), which suggests a decreased probability of glutamate release. Furthermore, the changes in the paired pulse ratio and in BK channel activity were blocked by paxilline, a BK channel blocker. Finally, we found that thalidomide-induced cognitive dysfunctions were restored by paxilline treatment. These results suggest that thalidomide-mediated BK channel hyperfunction is responsible for the pathological mechanism of thalidomide-associated reversible memory loss.


Assuntos
Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Imunossupressores/efeitos adversos , Indóis/uso terapêutico , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/uso terapêutico , Talidomida/efeitos adversos , Animais , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp
9.
J Neurosci ; 38(14): 3571-3583, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29530986

RESUMO

Mutations in the cereblon (CRBN) gene cause human intellectual disability, one of the most common cognitive disorders. However, the molecular mechanisms of CRBN-related intellectual disability remain poorly understood. We investigated the role of CRBN in synaptic function and animal behavior using male mouse and Drosophila models. Crbn knock-out (KO) mice showed normal brain and spine morphology as well as intact synaptic plasticity; however, they also exhibited decreases in synaptic transmission and presynaptic release probability exclusively in excitatory synapses. Presynaptic function was impaired not only by loss of CRBN expression, but also by expression of pathogenic CRBN mutants (human R419X mutant and Drosophila G552X mutant). We found that the BK channel blockers paxilline and iberiotoxin reversed this decrease in presynaptic release probability in Crbn KO mice. In addition, paxilline treatment also restored normal cognitive behavior in Crbn KO mice. These results strongly suggest that increased BK channel activity is the pathological mechanism of intellectual disability in CRBN mutations.SIGNIFICANCE STATEMENTCereblon (CRBN), a well known target of the immunomodulatory drug thalidomide, was originally identified as a gene that causes human intellectual disability when mutated. However, the molecular mechanisms of CRBN-related intellectual disability remain poorly understood. Based on the idea that synaptic abnormalities are the most common factor in cognitive dysfunction, we monitored the synaptic structure and function of Crbn knock-out (KO) animals to identify the molecular mechanisms of intellectual disability. Here, we found that Crbn KO animals showed cognitive deficits caused by enhanced BK channel activity and reduced presynaptic glutamate release. Our findings suggest a physiological pathomechanism of the intellectual disability-related gene CRBN and will contribute to the development of therapeutic strategies for CRBN-related intellectual disability.


Assuntos
Cognição , Deficiência Intelectual/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transmissão Sináptica , Proteínas Adaptadoras de Transdução de Sinal , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Drosophila , Ácido Glutâmico/metabolismo , Indóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/fisiologia
10.
J Exp Med ; 214(10): 2947-2966, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28899869

RESUMO

Vaccinia-related kinases (VRKs) are multifaceted serine/threonine kinases that play essential roles in various aspects of cell signaling, cell cycle progression, apoptosis, and neuronal development and differentiation. However, the neuronal function of VRK3 is still unknown despite its etiological potential in human autism spectrum disorder (ASD). Here, we report that VRK3-deficient mice exhibit typical symptoms of autism-like behavior, including hyperactivity, stereotyped behaviors, reduced social interaction, and impaired context-dependent spatial memory. A significant decrease in dendritic spine number and arborization were identified in the hippocampus CA1 of VRK3-deficient mice. These mice also exhibited a reduced rectification of AMPA receptor-mediated current and changes in expression of synaptic and signaling proteins, including tyrosine receptor kinase B (TrkB), Arc, and CaMKIIα. Notably, TrkB stimulation with 7,8-dihydroxyflavone reversed the altered synaptic structure and function and successfully restored autism-like behavior in VRK3-deficient mice. These results reveal that VRK3 plays a critical role in neurodevelopmental disorders and suggest a potential therapeutic strategy for ASD.


Assuntos
Transtorno Autístico/etiologia , Proteínas Serina-Treonina Quinases/deficiência , Receptor trkB/fisiologia , Animais , Região CA1 Hipocampal/patologia , Feminino , Flavanonas/farmacologia , Hipercinese/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/metabolismo , Receptor trkB/efeitos dos fármacos , Receptor trkB/metabolismo , Comportamento Social , Comportamento Estereotipado
11.
Front Mol Neurosci ; 10: 81, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28381988

RESUMO

Copy number variants and point mutations of NEPH2 (also called KIRREL3) gene encoding an immunoglobulin (Ig) superfamily adhesion molecule have been linked to autism spectrum disorders, intellectual disability and neurocognitive delay associated with Jacobsen syndrome, but the physiological roles of Neph2 in the mammalian brain remain largely unknown. Neph2 is highly expressed in the dentate granule (DG) neurons of the hippocampus and is localized in both dendrites and axons. It was recently shown that Neph2 is required for the formation of mossy fiber filopodia, the axon terminal structure of DG neurons forming synapses with GABAergic neurons of CA3. In contrast, however, it is unknown whether Neph2 also has any roles in the postsynaptic compartments of DG neurons. We here report that, through its C-terminal PDZ domain-binding motif, Neph2 directly interacts with postsynaptic density (PSD)-95, an abundant excitatory postsynaptic scaffolding protein. Moreover, Neph2 protein is detected in the brain PSD fraction and interacts with PSD-95 in synaptosomal lysates. Functionally, loss of Neph2 in mice leads to age-specific defects in the synaptic connectivity of DG neurons. Specifically, Neph2-/- mice show significantly increased spontaneous excitatory synaptic events in DG neurons at postnatal week 2 when the endogenous Neph2 protein expression peaks, but show normal excitatory synaptic transmission at postnatal week 3. The evoked excitatory synaptic transmission and synaptic plasticity of medial perforant pathway (MPP)-DG synapses are also normal in Neph2-/- mice at postnatal week 3, further confirming the age-specific synaptic defects. Together, our results provide some evidence for the postsynaptic function of Neph2 in DG neurons during the early postnatal period, which might be implicated in neurodevelopmental and cognitive disorders caused by NEPH2 mutations.

12.
Nat Commun ; 7: 12328, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27480238

RESUMO

Synaptic adhesion molecules regulate various aspects of synapse development, function and plasticity. These functions mainly involve trans-synaptic interactions and positive regulations, whereas cis-interactions and negative regulation are less understood. Here we report that SALM4, a member of the SALM/Lrfn family of synaptic adhesion molecules, suppresses excitatory synapse development through cis inhibition of SALM3, another SALM family protein with synaptogenic activity. Salm4-mutant (Salm4(-/-)) mice show increased excitatory synapse numbers in the hippocampus. SALM4 cis-interacts with SALM3, inhibits trans-synaptic SALM3 interaction with presynaptic LAR family receptor tyrosine phosphatases and suppresses SALM3-dependent presynaptic differentiation. Importantly, deletion of Salm3 in Salm4(-/-) mice (Salm3(-/-); Salm4(-/-)) normalizes the increased excitatory synapse number. These results suggest that SALM4 negatively regulates excitatory synapses via cis inhibition of the trans-synaptic SALM3-LAR adhesion.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Diferenciação Celular/fisiologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Terminações Pré-Sinápticas/fisiologia , Células Piramidais/fisiologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Adesão Celular/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Proteínas do Tecido Nervoso , Moléculas de Adesão de Célula Nervosa/genética , Transmissão Sináptica/fisiologia
13.
Cell Rep ; 14(4): 808-822, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26776509

RESUMO

The four members of the LRRTM family (LRRTM1-4) are postsynaptic adhesion molecules essential for excitatory synapse development. They have also been implicated in neuropsychiatric diseases. Here, we focus on LRRTM3, showing that two distinct LRRTM3 variants generated by alternative splicing regulate LRRTM3 interaction with PSD-95, but not its excitatory synapse-promoting activity. Overexpression of either LRRTM3 variant increased excitatory synapse density in dentate gyrus (DG) granule neurons, whereas LRRTM3 knockdown decreased it. LRRTM3 also controlled activity-regulated AMPA receptor surface expression in an alternative splicing-dependent manner. Furthermore, Lrrtm3-knockout mice displayed specific alterations in excitatory synapse density, excitatory synaptic transmission and excitability in DG granule neurons but not in CA1 pyramidal neurons. Lastly, LRRTM3 required only specific splice variants of presynaptic neurexins for their synaptogenic activity. Collectively, our data highlight alternative splicing and differential presynaptic ligand utilization in the regulation of LRRTMs, revealing key regulatory mechanisms for excitatory synapse development.


Assuntos
Processamento Alternativo , Moléculas de Adesão Celular Neuronais/metabolismo , Potenciais Pós-Sinápticos Excitadores , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/crescimento & desenvolvimento , Região CA1 Hipocampal/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Giro Denteado/citologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Transporte Proteico , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Ratos , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia
14.
Neurobiol Dis ; 87: 19-28, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26704708

RESUMO

In neurodegenerative diseases like AD, tau forms neurofibrillary tangles, composed of tau protein. In the AD brain, activated caspases cleave tau at the 421th Asp, generating a caspase-cleaved form of tau, TauC3. Although TauC3 is known to assemble rapidly into filaments in vitro, a role of TauC3 in vivo remains unclear. Here, we generated a transgenic mouse expressing human TauC3 using a neuron-specific promoter. In this mouse, we found that human TauC3 was expressed in the hippocampus and cortex. Interestingly, TauC3 mice showed drastic learning and spatial memory deficits and reduced synaptic density at a young age (2-3months). Notably, tau oligomers as well as tau aggregates were found in TauC3 mice showing memory deficits. Further, i.p. or i.c.v. injection with methylene blue or Congo red, inhibitors of tau aggregation in vitro, and i.p. injection with rapamycin significantly reduced the amounts of tau oligomers in the hippocampus, rescued spine density, and attenuated memory impairment in TauC3 mice. Together, these results suggest that TauC3 facilitates early memory impairment in transgenic mice accompanied with tau oligomer formation, providing insight into the role of TauC3 in the AD pathogenesis associated with tau oligomers and a useful AD model to test drug candidates.


Assuntos
Caspases/metabolismo , Transtornos da Memória/metabolismo , Proteínas tau/metabolismo , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/patologia , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nootrópicos/farmacologia , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/fisiologia , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Sirolimo/farmacologia , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Proteínas tau/genética
15.
Cell Rep ; 12(10): 1618-30, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26321637

RESUMO

Synaptic adhesion molecules regulate diverse aspects of synapse development and plasticity. SALM3 is a PSD-95-interacting synaptic adhesion molecule known to induce presynaptic differentiation in contacting axons, but little is known about its presynaptic receptors and in vivo functions. Here, we identify an interaction between SALM3 and LAR family receptor protein tyrosine phosphatases (LAR-RPTPs) that requires the mini-exon B splice insert in LAR-RPTPs. In addition, SALM3-dependent presynaptic differentiation requires all three types of LAR-RPTPs. SALM3 mutant (Salm3(-/-)) mice display markedly reduced excitatory synapse number but normal synaptic plasticity in the hippocampal CA1 region. Salm3(-/-) mice exhibit hypoactivity in both novel and familiar environments but perform normally in learning and memory tests administered. These results suggest that SALM3 regulates excitatory synapse development and locomotion behavior.


Assuntos
Moléculas de Adesão de Célula Nervosa/fisiologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Sinapses/fisiologia , Processamento Alternativo , Animais , Diferenciação Celular , Potenciais Pós-Sinápticos Excitadores , Éxons , Hipocampo/citologia , Hipocampo/metabolismo , Aprendizagem , Locomoção , Glicoproteínas de Membrana , Camundongos Knockout , Proteínas do Tecido Nervoso , Plasticidade Neuronal , Isoformas de Proteínas/fisiologia , Desempenho Psicomotor , Sítios de Splice de RNA , Transmissão Sináptica
16.
J Neurochem ; 132(3): 276-85, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25418874

RESUMO

Methyl-ß-cyclodextrin (MßCD) is a reagent that depletes cholesterol and disrupts lipid rafts, a type of cholesterol-enriched cell membrane microdomain. Lipid rafts are essential for neuronal functions such as synaptic transmission and plasticity, which are sensitive to even low doses of MßCD. However, how MßCD changes synaptic function, such as N-methyl-d-aspartate receptor (NMDA-R) activity, remains unclear. We monitored changes in synaptic transmission and plasticity after disrupting lipid rafts with MßCD. At low concentrations (0.5 mg/mL), MßCD decreased basal synaptic transmission and miniature excitatory post-synaptic current without changing NMDA-R-mediated synaptic transmission and the paired-pulse facilitation ratio. Interestingly, low doses of MßCD failed to deplete cholesterol or affect α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA-R) and NMDA-R levels, while clearly reducing GluA1 levels selectively in the synaptosomal fraction. Low doses of MßCD decreased the inhibitory effects of NASPM, an inhibitor for GluA2-lacking AMPA-R. MßCD successfully decreased NMDA-R-mediated long-term potentiation but did not affect the formation of either NMDA-R-mediated or group I metabotropic glutamate receptor-dependent long-term depression. MßCD inhibited de-depression without affecting de-potentiation. These results suggest that MßCD regulates GluA1-dependent synaptic potentiation but not synaptic depression in a cholesterol-independent manner.


Assuntos
Receptores de AMPA/fisiologia , Sinapses/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia , Animais , Colesterol/metabolismo , Técnicas In Vitro , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos
17.
Brain Res ; 1557: 34-42, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24560601

RESUMO

Melatonin is involved in various neuronal functions such as circadian rhythmicity and thermoregulation. Melatonin has a wide range of pharmacologically effective concentration levels from the nanomolar to millimolar levels. Recently, the antiepileptic effect of high dose melatonin has been the focus of clinical studies; however, its detailed mechanism especially in relation to neurotransmitter release and synaptic transmission remains unclear. We studied the effect of melatonin at high concentrations on the neurotransmitter release by monitoring norepinephrine release in PC12 cells, and excitatory postsynaptic potential in rat hippocampal slices. Melatonin inhibits the 70mM K(+)-induced Ca(2+) increase at millimolar levels without effect on bradykinin-triggered Ca(2+) increase in PC12 cells. Melatonin (1mM) did not affect A2A adenosine receptor-evoked cAMP production, and classical melatonin receptor antagonists did not reverse the melatonin-induced inhibitory effect, suggesting G-protein coupled receptor independency. Melatonin inhibits the 70mM K(+)-induced norepinephrine release at a similar effective concentration range in PC12 cells. We confirmed that melatonin (100µM) inhibits excitatory synaptic transmission of the hippocampal Schaffer collateral pathway with the decrease in basal synaptic transmission and the increase in paired pulse ratio. These results show that melatonin inhibits neurotransmitter release through the blocking of voltage-sensitive Ca(2+) channels and suggest a possible mechanism for the antiepileptic effect of melatonin.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Hipocampo/efeitos dos fármacos , Melatonina/farmacologia , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , AMP Cíclico/metabolismo , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Células HEK293 , Hipocampo/fisiologia , Humanos , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Norepinefrina/metabolismo , Células PC12 , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina/metabolismo , Receptores de Melatonina/agonistas , Receptores de Melatonina/antagonistas & inibidores , Receptores de Melatonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
18.
J Clin Invest ; 123(7): 2791-802, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23921129

RESUMO

Amyloid-ß (Aß) induces neuronal loss and cognitive deficits and is believed to be a prominent cause of Alzheimer's disease (AD); however, the cellular pathology of the disease is not fully understood. Here, we report that IgG Fcγ receptor II-b (FcγRIIb) mediates Aß neurotoxicity and neurodegeneration. We found that FcγRIIb is significantly upregulated in the hippocampus of AD brains and neuronal cells exposed to synthetic Aß. Neuronal FcγRIIb activated ER stress and caspase-12, and Fcgr2b KO primary neurons were resistant to synthetic Aß-induced cell death in vitro. Fcgr2b deficiency ameliorated Aß-induced inhibition of long-term potentiation and inhibited the reduction of synaptic density by naturally secreted Aß. Moreover, genetic depletion of Fcgr2b rescued memory impairments in an AD mouse model. To determine the mechanism of action of FcγRIIb in Aß neurotoxicity, we demonstrated that soluble Aß oligomers interact with FcγRIIb in vitro and in AD brains, and that inhibition of their interaction blocks synthetic Aß neurotoxicity. We conclude that FcγRIIb has an aberrant, but essential, role in Aß-mediated neuronal dysfunction.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/fisiologia , Transtornos da Memória/metabolismo , Fragmentos de Peptídeos/fisiologia , Receptores de IgG/fisiologia , Doença de Alzheimer/patologia , Amiloide/fisiologia , Animais , Células CHO , Cricetinae , Potenciais Pós-Sinápticos Excitadores , Feminino , Hipocampo/patologia , Humanos , Masculino , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Cultura Primária de Células , Transdução de Sinais , Sinapses/fisiologia , Ativação Transcricional
19.
Nat Commun ; 4: 1579, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23481387

RESUMO

Dopaminergic systems are implicated in stress-related behaviour. Here we investigate behavioural responses to chronic stress in dopamine D2 receptor knockout mice and find that anxiety-like behaviours are increased compared with wild-type mice. Repeated stress exposure suppresses cocaine-induced behavioural sensitization, cocaine-seeking and relapse behaviours in dopamine D2 receptor knockout mice. Cocaine challenge after drug withdrawal in cocaine-experienced wild-type or dopamine D2 receptor knockout mice is associated with inhibition of long-term depression in the nucleus accumbens, and chronic stress during withdrawal prevents inhibition after cocaine challenge in cocaine-experienced dopamine D2 receptor knockout mice, but not in wild-type mice. Lentiviral-induced knockdown of dopamine D2 receptors in the nucleus accumbens of wild-type mice does not affect basal locomotor activity, but confers stress-induced inhibition of the expression of cocaine-induced behavioural sensitization. Stressed mice depleted of dopamine D2 receptors do not manifest long-term depression inhibition. Our results suggest that dopamine D2 receptors have roles in regulating synaptic modification triggered by stress and drug addiction.


Assuntos
Comportamento Aditivo/metabolismo , Comportamento Aditivo/fisiopatologia , Comportamento Animal , Plasticidade Neuronal , Receptores de Dopamina D2/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Animais , Ansiedade/complicações , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Comportamento Aditivo/etiologia , Comportamento Animal/efeitos dos fármacos , Sensibilização do Sistema Nervoso Central/efeitos dos fármacos , Cocaína/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Estimulação Elétrica , Técnicas de Silenciamento de Genes , Técnicas In Vitro , Lentivirus/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/patologia , Núcleo Accumbens/fisiopatologia , Receptores de Dopamina D2/deficiência , Restrição Física , Estresse Psicológico/complicações
20.
Chin J Integr Med ; 17(8): 575-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21826591

RESUMO

OBJECTIVE: Moxibustion is currently used for treating diabetes mellitus (DM) as a non-drug intervention in East Asian countries. This systematic review aims to evaluate the effectiveness of moxibustion for managing the symptoms of type 2 DM patients. METHODS: We searched MEDLINE, AMED, EMBASE, CINAHL, The Cochrane Library, six Korean databases, and four Chinese databases. Risk of bias was used for evaluating the quality of the included studies. RESULTS: A total of 5 studies met the inclusion criteria for this review. All of the included studies had high risks of bias. One randomized clinical trial (RCT) compared the effectiveness of one-time moxibustion use with oral administration of glibenclimide and showed the significant effects of moxibustion on glycemic control. Another RCT tested the effectiveness of moxibustion plus conventional treatment, and the moxibustion group reported significant improvement in fasting and postprandial blood glucose levels compared with the conventional treatment group. Two RCTs compared the effectiveness of moxibustion versus acupuncture or moxibustion plus acupuncture, and the combined treatment showed the most favorable effects on the control of blood glucose, urine glucose, and glycocylated hemoglobin A(1c) (HbA(1c)). One uncontrolled observational study showed favorable effects of moxibustion on the response rate. CONCLUSIONS: It is difficult to conclude that moxibustion is an effective intervention for the control of type 2 DM due to the scarcity of trials and the low methodological quality of included studies. Further rigorous RCTs may be necessary to evaluate the effectiveness of moxibustion for type 2 DM.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Moxibustão/métodos , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA