Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 9(43): eadj1010, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37878694

RESUMEN

The time of day strongly influences adaptive behaviors like long-term memory, but the correlating synaptic and molecular mechanisms remain unclear. The circadian clock comprises a canonical transcription-translation feedback loop (TTFL) strictly dependent on the BMAL1 transcription factor. We report that BMAL1 rhythmically localizes to hippocampal synapses in a manner dependent on its phosphorylation at Ser42 [pBMAL1(S42)]. pBMAL1(S42) regulates the autophosphorylation of synaptic CaMKIIα and circadian rhythms of CaMKIIα-dependent molecular interactions and LTP but not global rest/activity behavior. Therefore, our results suggest a model in which repurposing of the clock protein BMAL1 to synapses locally gates the circadian timing of plasticity.


Asunto(s)
Factores de Transcripción ARNTL , Relojes Circadianos , Fosforilación , Factores de Transcripción ARNTL/genética , Ritmo Circadiano/fisiología , Hipocampo/metabolismo
2.
Cell ; 186(18): 3845-3861.e24, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37591240

RESUMEN

Dopaminergic projections regulate various brain functions and are implicated in many neuropsychiatric disorders. There are two anatomically and functionally distinct dopaminergic projections connecting the midbrain to striatum: nigrostriatal, which controls movement, and mesolimbic, which regulates motivation. However, how these discrete dopaminergic synaptic connections are established is unknown. Through an unbiased search, we identify that two groups of antagonistic TGF-ß family members, bone morphogenetic protein (BMP)6/BMP2 and transforming growth factor (TGF)-ß2, regulate dopaminergic synapse development of nigrostriatal and mesolimbic neurons, respectively. Projection-preferential expression of their receptors contributes to specific synapse development. Downstream, Smad1 and Smad2 are specifically activated and required for dopaminergic synapse development and function in nigrostriatal vs. mesolimbic projections. Remarkably, Smad1 mutant mice show motor defects, whereas Smad2 mutant mice show lack of motivation. These results uncover the molecular logic underlying the proper establishment of functionally segregated dopaminergic synapses and may provide strategies to treat relevant, projection-specific disease symptoms by targeting specific BMPs/TGF-ß and/or Smads.


Asunto(s)
Cuerpo Estriado , Dopamina , Animales , Ratones , Mesencéfalo , Motivación , Movimiento , Sinapsis
3.
Neuroscientist ; : 10738584231170167, 2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37140155

RESUMEN

The refinement of immature neuronal networks into efficient mature ones is critical to nervous system development and function. This process of synapse refinement is driven by the neuronal activity-dependent competition of converging synaptic inputs, resulting in the elimination of weak inputs and the stabilization of strong ones. Neuronal activity, whether in the form of spontaneous activity or experience-evoked activity, is known to drive synapse refinement in numerous brain regions. More recent studies are now revealing the manner and mechanisms by which neuronal activity is detected and converted into molecular signals that appropriately regulate the elimination of weaker synapses and stabilization of stronger ones. Here, we highlight how spontaneous activity and evoked activity instruct neuronal activity-dependent competition during synapse refinement. We then focus on how neuronal activity is transformed into the molecular cues that determine and execute synapse refinement. A comprehensive understanding of the mechanisms underlying synapse refinement can lead to novel therapeutic strategies in neuropsychiatric diseases characterized by aberrant synaptic function.

4.
Mol Psychiatry ; 2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36737483

RESUMEN

Functional and structural connectivity alterations in short- and long-range projections have been reported across neurodevelopmental disorders (NDD). Interhemispheric callosal projection neurons (CPN) represent one of the major long-range projections in the brain, which are particularly important for higher-order cognitive function and flexibility. However, whether a causal relationship exists between interhemispheric connectivity alterations and cognitive deficits in NDD remains elusive. Here, we focused on CDKL5 Deficiency Disorder (CDD), a severe neurodevelopmental disorder caused by mutations in the X-linked Cyclin-dependent kinase-like 5 (CDKL5) gene. We found an increase in homotopic interhemispheric connectivity and functional hyperconnectivity across higher cognitive areas in adult male and female CDKL5-deficient mice by resting-state functional MRI (rs-fMRI) analysis. This was accompanied by an increase in the number of callosal synaptic inputs but decrease in local synaptic connectivity in the cingulate cortex of juvenile CDKL5-deficient mice, suggesting an impairment in excitatory synapse development and a differential role of CDKL5 across excitatory neuron subtypes. These deficits were associated with significant cognitive impairments in CDKL5 KO mice. Selective deletion of CDKL5 in the largest subtype of CPN likewise resulted in an increase of functional callosal inputs, without however significantly altering intracortical cingulate networks. Notably, such callosal-specific changes were sufficient to cause cognitive deficits. Finally, when CDKL5 was selectively re-expressed only in this CPN subtype, in otherwise CDKL5-deficient mice, it was sufficient to prevent the cognitive impairments of CDKL5 mutants. Together, these results reveal a novel role of CDKL5 by demonstrating that it is both necessary and sufficient for proper CPN connectivity and cognitive function and flexibility, and further validates a causal relationship between CPN dysfunction and cognitive impairment in a model of NDD.

5.
Curr Opin Neurobiol ; 79: 102692, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36805716

RESUMEN

The formation of appropriate synaptic connections is critical for the proper functioning of the brain. Early in development, neurons form a surplus of immature synapses. To establish efficient, functional neural networks, neurons selectively stabilize active synapses and eliminate less active ones. This process is known as activity-dependent synapse refinement. Defects in this process have been implicated in neuropsychiatric disorders such as schizophrenia and autism. Here we review the manner and mechanisms by which synapse elimination is regulated through activity-dependent competition. We propose a theoretical framework for the molecular mechanisms of synapse refinement, in which three types of signals regulate the refinement. We then describe the identity of these signals and discuss how multiple molecular signals interact to achieve appropriate synapse refinement in the brain.


Asunto(s)
Neuronas , Sinapsis , Neuronas/fisiología , Sinapsis/fisiología , Encéfalo
6.
J Neurosci ; 42(21): 4250-4266, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35504727

RESUMEN

The Protocadherin-10 (PCDH10) gene is associated with autism spectrum disorder (ASD), obsessive-compulsive disorder (OCD), and major depression (MD). The PCDH10 protein is a homophilic cell adhesion molecule that belongs to the δ2-protocadherin family. PCDH10 is highly expressed in the developing brain, especially in the basolateral nucleus of the amygdala (BLA). However, the role of PCDH10 in vivo has been debatable: one paper reported that a Pcdh10 mutant mouse line showed changes in axonal projections; however, another Pcdh10 mutant mouse line was reported to have failed to detect axonal phenotypes. Therefore, the actual roles of PCDH10 in the brain remain to be elucidated. We established a new Pcdh10 KO mouse line using the CRISPR/Cas9 system, without inserting gene cassettes to avoid nonspecific effects, examined the roles of PCDH10 in the brain, and studied the behavioral consequences of Pcdh10 inactivation. Here, we show that Pcdh10 KO mice do not show defects in axonal development. Instead, we find that Pcdh10 KO mice exhibit impaired development of excitatory synapses in the dorsal BLA. We further demonstrate that male Pcdh10 KO mice exhibit reduced anxiety-related behaviors, impaired fear conditioning, decreased stress-coping responses, and mildly impaired social recognition and communication. These results indicate that PCDH10 plays a critical role in excitatory synapse development, but not axon development, in the dorsal BLA and that PCDH10 regulates anxiety-related, fear-related, and stress-related behaviors. Our results reveal the roles of PCDH10 in the brain and its relationship to relevant psychiatric disorders such as ASD, OCD, and MD.SIGNIFICANCE STATEMENTProtocadherin-10 (PCDH10) encodes a cell adhesion molecule and is implicated in autism spectrum disorder (ASD), obsessive-compulsive disorder (OCD), and major depression (MD). PCDH10 is highly expressed in the basolateral nucleus of the amygdala (BLA). However, the phenotypes of previously published Pcdh10 mutant mice are debatable, and some are possibly because of the nonspecific effects of the LacZ/Neo cassette inserted in the mice. We have generated a new Pcdh10 mutant mouse line without the LacZ/Neo cassette. Using our new mouse line, we reveal the roles of PCDH10 for excitatory synapse development in the BLA. The mutant mice exhibit anxiety-related, fear-related, and stress-related behaviors, which are relevant to ASD, OCD, and MD, suggesting a possible treatment strategy for such psychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Obsesivo Compulsivo , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Ansiedad/psicología , Trastorno del Espectro Autista/metabolismo , Miedo/fisiología , Humanos , Masculino , Ratones , Protocadherinas , Sinapsis/metabolismo
7.
STAR Protoc ; 2(3): 100742, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34430915

RESUMEN

The visual system is the best system to study activity-dependent sensory circuit development. The connections from the retina to the dorsal lateral geniculate nucleus, the retinogeniculate connections, undergo extensive remodeling during early postnatal life. Thus, techniques that allow the expression of transgenes early in the developing retina are essential to study visual system development. Here, we describe a protocol to express genes-of-interest in the developing mouse retina via in utero intraocular adeno-associated virus injections. For complete details on the use and execution of this protocol, please refer to Yasuda et al. (2021).


Asunto(s)
Inyecciones Intraoculares/métodos , Retina/embriología , Transgenes/genética , Animales , Dependovirus/genética , Feto/cirugía , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Ratones/embriología , Retina/crecimiento & desarrollo , Sinapsis , Transcriptoma/genética , Vías Visuales/crecimiento & desarrollo
8.
Science ; 372(6539)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33859005

RESUMEN

Protocadherin-19 (PCDH19) mutations cause early-onset seizures and cognitive impairment. The PCDH19 gene is on the X-chromosome. Unlike most X-linked disorders, PCDH19 mutations affect heterozygous females (PCDH19HET♀ ) but not hemizygous males (PCDH19HEMI♂ ); however, the reason why remains to be elucidated. We demonstrate that PCDH19, a cell-adhesion molecule, is enriched at hippocampal mossy fiber synapses. Pcdh19HET♀ but not Pcdh19HEMI♂ mice show impaired mossy fiber synaptic structure and physiology. Consistently, Pcdh19HET♀ but not Pcdh19HEMI♂ mice exhibit reduced pattern completion and separation abilities, which require mossy fiber synaptic function. Furthermore, PCDH19 appears to interact with N-cadherin at mossy fiber synapses. In Pcdh19HET♀ conditions, mismatch between PCDH19 and N-cadherin diminishes N-cadherin-dependent signaling and impairs mossy fiber synapse development; N-cadherin overexpression rescues Pcdh19HET♀ phenotypes. These results reveal previously unknown molecular and cellular mechanisms underlying the female-specific PCDH19 disorder phenotype.


Asunto(s)
Cadherinas/metabolismo , Disfunción Cognitiva/fisiopatología , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Fibras Musgosas del Hipocampo/fisiopatología , Sinapsis/fisiología , Animales , Región CA3 Hipocampal/fisiopatología , Región CA3 Hipocampal/ultraestructura , Cadherinas/genética , Disfunción Cognitiva/genética , Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/fisiopatología , Femenino , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Potenciación a Largo Plazo , Masculino , Ratones , Fibras Musgosas del Hipocampo/ultraestructura , Mutación , Protocadherinas , Caracteres Sexuales , Sinapsis/ultraestructura , beta Catenina/metabolismo
9.
Neuron ; 109(8): 1333-1349.e6, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33770504

RESUMEN

To establish functional neural circuits in the brain, synaptic connections are refined by neural activity during development, where active connections are maintained and inactive ones are eliminated. However, the molecular signals that regulate synapse refinement remain to be elucidated. When we inactivate a subset of neurons in the mouse cingulate cortex, their callosal connections are eliminated through activity-dependent competition. Using this system, we identify JAK2 tyrosine kinase as a key regulator of inactive synapse elimination. We show that JAK2 is necessary and sufficient for elimination of inactive connections; JAK2 is activated at inactive synapses in response to signals from other active synapses; STAT1, a substrate of JAK2, mediates inactive synapse elimination; JAK2 signaling is critical for physiological refinement of synapses during normal development; and JAK2 regulates synapse refinement in multiple brain regions. We propose that JAK2 is an activity-dependent switch that serves as a determinant of inactive synapse elimination.


Asunto(s)
Giro del Cíngulo/fisiología , Janus Quinasa 2/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Giro del Cíngulo/metabolismo , Ratones , Neuronas/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Sinapsis/metabolismo
10.
EMBO J ; 39(16): e104136, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32452062

RESUMEN

Developmental synaptic remodeling is important for the formation of precise neural circuitry, and its disruption has been linked to neurodevelopmental disorders such as autism and schizophrenia. Microglia prune synapses, but integration of this synapse pruning with overlapping and concurrent neurodevelopmental processes, remains elusive. Adhesion G protein-coupled receptor ADGRG1/GPR56 controls multiple aspects of brain development in a cell type-specific manner: In neural progenitor cells, GPR56 regulates cortical lamination, whereas in oligodendrocyte progenitor cells, GPR56 controls developmental myelination and myelin repair. Here, we show that microglial GPR56 maintains appropriate synaptic numbers in several brain regions in a time- and circuit-dependent fashion. Phosphatidylserine (PS) on presynaptic elements binds GPR56 in a domain-specific manner, and microglia-specific deletion of Gpr56 leads to increased synapses as a result of reduced microglial engulfment of PS+ presynaptic inputs. Remarkably, a particular alternatively spliced isoform of GPR56 is selectively required for microglia-mediated synaptic pruning. Our present data provide a ligand- and isoform-specific mechanism underlying microglial GPR56-mediated synapse pruning in the context of complex neurodevelopmental processes.


Asunto(s)
Empalme Alternativo , Microglía/metabolismo , Fosfatidilserinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sinapsis/metabolismo , Animales , Ratones , Ratones Transgénicos , Microglía/citología , Fosfatidilserinas/genética , Unión Proteica , Isoformas de Proteínas , Receptores Acoplados a Proteínas G/genética , Sinapsis/genética
11.
Neuroreport ; 31(2): 125-130, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31895747

RESUMEN

Growth factor signaling in the brain is implicated in many neuropsychiatric disorders, including depression, autism, and epilepsy. Fibroblast growth factor 22 is a growth factor that regulates excitatory synapse development and neurogenesis in the brain. We have previously shown that adult mice in which fibroblast growth factor 22 is constitutively inactivated in all cells throughout life (fibroblast growth factor 22-null mice) show anhedonia, a core feature of depression in humans, suggesting that fibroblast growth factor 22 signaling contributes to the regulation of affective behavior. Here we asked (1) whether inactivation of fibroblast growth factor 22 specifically in neurons is sufficient to induce anhedonia in mice and (2) whether fibroblast growth factor 22 signaling is important during development or in adults for the regulation of affective behavior. To address these questions, we performed the sucrose preference test, which is used as an indicator of anhedonia, with neuron-specific conditional fibroblast growth factor 22 knockout mice, in which fibroblast growth factor 22 is inactivated in neurons at birth (neonatal-fibroblast growth factor 22-knockout mice) or in adults (adult-fibroblast growth factor 22-knockout mice). We found that neonatal-fibroblast growth factor 22-knockout mice show anhedonia (decreased preference for sucrose), while adult-fibroblast growth factor 22-knockout mice do not. Therefore, neuronal fibroblast growth factor 22 signaling is critical during development, and not in adults, for the regulation of affective behavior. Our work also implies that defects in growth factor-dependent synapse development, neurogenesis, or both may underlie depression of a developmental origin.


Asunto(s)
Anhedonia/fisiología , Encéfalo/crecimiento & desarrollo , Factores de Crecimiento de Fibroblastos/genética , Neurogénesis/genética , Envejecimiento , Animales , Encéfalo/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Ratones Transgénicos , Neurogénesis/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología , Sinapsis/metabolismo
12.
Neuron ; 100(1): 120-134.e6, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30308165

RESUMEN

Microglia regulate synaptic circuit remodeling and phagocytose synaptic material in the healthy brain; however, the mechanisms directing microglia to engulf specific synapses and avoid others remain unknown. Here, we demonstrate that an innate immune signaling pathway protects synapses from inappropriate removal. The expression patterns of CD47 and its receptor, SIRPα, correlated with peak pruning in the developing retinogeniculate system, and mice lacking these proteins exhibited increased microglial engulfment of retinogeniculate inputs and reduced synapse numbers in the dorsal lateral geniculate nucleus. CD47-deficient mice also displayed increased functional pruning, as measured by electrophysiology. In addition, CD47 was found to be required for neuronal activity-mediated changes in engulfment, as microglia in CD47 knockout mice failed to display preferential engulfment of less active inputs. Taken together, these results demonstrate that CD47-SIRPα signaling prevents excess microglial phagocytosis and show that molecular brakes can be regulated by activity to protect specific inputs.


Asunto(s)
Antígeno CD47/metabolismo , Microglía/metabolismo , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Sinapsis/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/fisiología , Receptores Inmunológicos/metabolismo
13.
J Biol Chem ; 293(31): 12026-12042, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-29914984

RESUMEN

Synapse maturation is a neural activity-dependent process during brain development, in which active synapses preferentially undergo maturation to establish efficient neural circuits in the brain. Defects in this process are implicated in various neuropsychiatric disorders. We have previously reported that a postsynaptic transmembrane protein, signal regulatory protein-α (SIRPα), plays an important role in activity-dependently directing synapse maturation. In the presence of synaptic activity, the ectodomain of SIRPα is cleaved and released and then acts as a retrograde signal to induce presynaptic maturation. However, how SIRPα detects synaptic activity to promote its ectodomain cleavage and synapse maturation is unknown. Here, we show that activity-dependent tyrosine phosphorylation of SIRPα is critical for SIRPα cleavage and synapse maturation. We found that during synapse maturation and in response to neural activity, SIRPα is highly phosphorylated on its tyrosine residues in the hippocampus, a structure critical for learning and memory. Tyrosine phosphorylation of SIRPα was necessary for SIRPα cleavage and presynaptic maturation, as indicated by the fact that a phosphorylation-deficient SIRPα variant underwent much less cleavage and could not drive presynaptic maturation. However, SIRPα phosphorylation did not affect its synaptic localization. Finally, we show that inhibitors of the Src and JAK kinase family suppress neural activity-dependent SIRPα phosphorylation and cleavage. Together, our results indicate that SIRPα phosphorylation serves as a mechanism for detecting synaptic activity and linking it to the ectodomain cleavage of SIRPα, which in turn drives synapse maturation in an activity-dependent manner.


Asunto(s)
Memoria/fisiología , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Receptores Inmunológicos/metabolismo , Sinapsis/metabolismo , Tirosina/metabolismo , Animales , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fosforilación , Cloruro de Potasio/farmacología , Cultivo Primario de Células , Dominios Proteicos , Proteolisis , Receptores Inmunológicos/genética , Sinapsis/efectos de los fármacos , Transmisión Sináptica , Inhibidores Tisulares de Metaloproteinasas/farmacología , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
14.
Neurosci Res ; 116: 60-69, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27965136

RESUMEN

Activity-dependent remodeling of neuronal connections is critical to nervous system development and function. These processes rely on the ability of synapses to detect neuronal activity and translate it into the appropriate molecular signals. One way to convert neuronal activity into downstream signaling is the proteolytic cleavage of cell adhesion molecules (CAMs). Here we review studies demonstrating the mechanisms by which proteolytic processing of CAMs direct the structural and functional remodeling of excitatory glutamatergic synapses during development and plasticity. Specifically, we examine how extracellular proteolytic cleavage of CAMs switches on or off molecular signals to 1) permit, drive, or restrict synaptic maturation during development and 2) strengthen or weaken synapses during adult plasticity. We will also examine emerging studies linking improper activity-dependent proteolytic processing of CAMs to neurological disorders such as schizophrenia, brain tumors, and Alzheimer's disease. Together these findings suggest that the regulation of activity-dependent proteolytic cleavage of CAMs is vital to proper brain development and lifelong function.


Asunto(s)
Encéfalo/fisiología , Moléculas de Adhesión Celular/metabolismo , Sinapsis/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Neoplasias Encefálicas/metabolismo , Humanos , Plasticidad Neuronal , Neuronas/metabolismo , Proteolisis , Esquizofrenia/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-29311892

RESUMEN

Various growth factors regulate synapse development and neurogenesis, and are essential for brain function. Changes in growth factor signaling are implicated in many neuropsychiatric disorders such as depression, autism and epilepsy. We have previously identified that fibroblast growth factor 22 (FGF22) is critical for excitatory synapse formation in several brain regions including the hippocampus. Mice with a genetic deletion of FGF22 (FGF22 null mice) have fewer excitatory synapses in the hippocampus. We have further found that as a behavioral consequence, FGF22 null mice show a depression-like behavior phenotype such as increased passive stress-coping behavior and anhedonia, without any changes in motor, anxiety, or social cognitive tests, suggesting that FGF22 is specifically important for affective behavior. Thus, addressing the precise roles of FGF22 in the brain will help understand how synaptogenic growth factors regulate affective behavior. In the hippocampus, FGF22 is expressed mainly by CA3 pyramidal neurons, but also by a subset of dentate granule cells. We find that in addition to synapse formation, FGF22 also contributes to neurogenesis in the dentate gyrus: FGF22 null mice show decreased dentate neurogenesis. To understand the cell type-specific roles of FGF22, we generated and analyzed CA3-specific FGF22 knockout mice (FGF22-CA3KO). We show that FGF22-CA3KO mice have reduced excitatory synapses on CA3 pyramidal neurons, but do not show changes in dentate neurogenesis. Behaviorally, FGF22-CA3KO mice still show increased immobility and decreased latency to float in the forced swim test and decreased preference for sucrose in the sucrose preference test, which are suggestive of a depressive-like phenotype similar to FGF22 null mice. These results demonstrate that: (i) CA3-derived FGF22 serves as a target-derived excitatory synaptic organizer in CA3 in vivo; (ii) FGF22 plays important roles in dentate neurogenesis, but CA3-derived FGF22 is not involved in neurogenesis; and (iii) a depression-like phenotype can result from FGF22 inactivation selectively in CA3 pyramidal neurons. Our results link the role of CA3-derived FGF22 in synapse development, and not in neurogenesis, to affective behavior.

16.
Elife ; 52016 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-28001126

RESUMEN

The SNARE-mediated vesicular transport pathway plays major roles in synaptic remodeling associated with formation of long-term memories, but the mechanisms that regulate this pathway during memory acquisition are not fully understood. Here we identify miRNAs that are up-regulated in the rodent hippocampus upon contextual fear-conditioning and identify the vesicular transport and synaptogenesis pathways as the major targets of the fear-induced miRNAs. We demonstrate that miR-153, a member of this group, inhibits the expression of key components of the vesicular transport machinery, and down-regulates Glutamate receptor A1 trafficking and neurotransmitter release. MiR-153 expression is specifically induced during LTP induction in hippocampal slices and its knockdown in the hippocampus of adult mice results in enhanced fear memory. Our results suggest that miR-153, and possibly other fear-induced miRNAs, act as components of a negative feedback loop that blocks neuronal hyperactivity at least partly through the inhibition of the vesicular transport pathway.


Asunto(s)
Miedo , Retroalimentación Fisiológica , Hipocampo/fisiología , Memoria , MicroARNs/metabolismo , Neuronas/fisiología , Vesículas Sinápticas/metabolismo , Animales , Ratones , Neurotransmisores/metabolismo , Receptores de Glutamato/metabolismo
17.
Neurogenesis (Austin) ; 3(1): e1168504, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27605441

RESUMEN

Brain development involves multiple levels of molecular coordination in forming a functional nervous system. The hippocampus is a brain area that is important for memory formation and spatial reasoning. During early postnatal development of the hippocampal circuit, Fibroblast growth factor 22 (FGF22) and FGF7 act to establish a balance of excitatory and inhibitory tone. Both FGFs are secreted from CA3 dendrites, acting on excitatory or inhibitory axon terminals formed onto CA3 dendrites, respectively. Mechanistically, FGF22 utilizes FGFR2b and FGFR1b to induce synaptic vesicle recruitment within axons of dentate granule cells (DGCs), and FGF7 utilizes FGFR2b to induce synaptic vesicle recruitment within interneuron axons. FGF signaling eventually induces gene expression in the presynaptic neurons; however, the effects of FGF22-induced gene expression within DGCs and FGF7-induced gene expression within interneurons in the context of a developing hippocampal circuit have yet to be explored. Here, we propose one hypothetical mechanism of FGF22-induced gene expression in controlling adult neurogenesis.

18.
Sci Rep ; 6: 33592, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27627962

RESUMEN

Functional synapse formation requires tight coordination between pre- and post-synaptic termini. Previous studies have shown that postsynaptic expression of heparan sulfate proteoglycan syndecan-2 (SDC2) induces dendritic spinogenesis. Those SDC2-induced dendritic spines are frequently associated with presynaptic termini. However, how postsynaptic SDC2 accelerates maturation of corresponding presynaptic termini is unknown. Because fibroblast growth factor 22 (FGF22), a heparan sulfate binding growth factor, has been shown to act as a presynaptic organizer released from the postsynaptic site, it seems possible that postsynaptic SDC2 presents FGF22 to the presynaptic FGF receptor to promote presynaptic differentiation. Here, we show that postsynaptic SDC2 uses its ectodomain to interact with and facilitate dendritic filopodial targeting of FGF22, triggering presynaptic maturation. Since SDC2 also enhances filopodial targeting of NMDAR via interaction with the CASK-mLIN7-MINT1 adaptor complex, presynaptic maturation promoted by FGF22 further feeds back to activate NMDAR at corresponding postsynaptic sites through increased neurotransmitter release and, consequently, promotes the dendritic filopodia-spines (F-S) transition. Meanwhile, via regulation of the KIF17 motor, CaMKII (activated by the NMDAR pathway) may further facilitate FGF22 targeting to dendritic filopodia that receive presynaptic stimulation. Our study suggests a positive feedback that promotes the coordination of postsynaptic and presynaptic differentiation.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Terminales Presinápticos/metabolismo , Transducción de Señal , Sindecano-2/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular Tumoral , Espinas Dendríticas/metabolismo , Heparitina Sulfato/metabolismo , Cinesinas , Ratones , Modelos Biológicos , Unión Proteica , Dominios Proteicos , Seudópodos/metabolismo , Ratas Sprague-Dawley , Sindecano-2/química
19.
Behav Brain Res ; 307: 11-7, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27036645

RESUMEN

Specific growth factors induce formation and differentiation of excitatory and inhibitory synapses, and are essential for brain development and function. Fibroblast growth factor 22 (FGF22) is important for specifying excitatory synapses during development, including in the hippocampus. Mice with a genetic deletion of FGF22 (FGF22KO) during development subsequently have fewer hippocampal excitatory synapses in adulthood. As a result, FGF22KO mice are resistant to epileptic seizure induction. In addition to playing a key role in learning, the hippocampus is known to mediate mood and anxiety. Here, we explored whether loss of FGF22 alters affective, anxiety or social cognitive behaviors in mice. We found that relative to control mice, FGF22KO mice display longer duration of floating and decreased latency to float in the forced swim test, increased immobility in the tail suspension test, and decreased preference for sucrose in the sucrose preference test, which are all suggestive of a depressive-like phenotype. No differences were observed between control and FGF22KO mice in other behavioral assays, including motor, anxiety, or social cognitive tests. These results suggest a novel role for FGF22 specifically in affective behaviors.


Asunto(s)
Depresión/genética , Depresión/fisiopatología , Factores de Crecimiento de Fibroblastos/deficiencia , Adaptación Ocular/genética , Animales , Cognición/fisiología , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Femenino , Factores de Crecimiento de Fibroblastos/genética , Preferencias Alimentarias/fisiología , Suspensión Trasera , Pérdida de Tono Postural/fisiología , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Actividad Motora/fisiología , Conducta Social , Sacarosa/administración & dosificación , Natación/fisiología , Natación/psicología
20.
Elife ; 52016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27083047

RESUMEN

Communication between pre- and postsynaptic cells promotes the initial organization of synaptic specializations, but subsequent synaptic stabilization requires transcriptional regulation. Here we show that fibroblast growth factor 22 (FGF22), a target-derived presynaptic organizer in the mouse hippocampus, induces the expression of insulin-like growth factor 2 (IGF2) for the stabilization of presynaptic terminals. FGF22 is released from CA3 pyramidal neurons and organizes the differentiation of excitatory nerve terminals formed onto them. Local application of FGF22 on the axons of dentate granule cells (DGCs), which are presynaptic to CA3 pyramidal neurons, induces IGF2 in the DGCs. IGF2, in turn, localizes to DGC presynaptic terminals and stabilizes them in an activity-dependent manner. IGF2 application rescues presynaptic defects of Fgf22(-/-) cultures. IGF2 is dispensable for the initial presynaptic differentiation, but is required for the following presynaptic stabilization both in vitro and in vivo. These results reveal a novel feedback signal that is critical for the activity-dependent stabilization of presynaptic terminals in the mammalian hippocampus.


Asunto(s)
Comunicación Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Hipocampo/fisiología , Factor II del Crecimiento Similar a la Insulina/biosíntesis , Neuronas/fisiología , Sinapsis/fisiología , Animales , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...