Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(6): 1450-1464.e23, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30100184

RESUMEN

Synapses are fundamental units of communication in the brain. The prototypical synapse-organizing complex neurexin-neuroligin mediates synapse development and function and is central to a shared genetic risk pathway in autism and schizophrenia. Neurexin's role in synapse development is thought to be mediated purely by its protein domains, but we reveal a requirement for a rare glycan modification. Mice lacking heparan sulfate (HS) on neurexin-1 show reduced survival, as well as structural and functional deficits at central synapses. HS directly binds postsynaptic partners neuroligins and LRRTMs, revealing a dual binding mode involving intrinsic glycan and protein domains for canonical synapse-organizing complexes. Neurexin HS chains also bind novel ligands, potentially expanding the neurexin interactome to hundreds of HS-binding proteins. Because HS structure is heterogeneous, our findings indicate an additional dimension to neurexin diversity, provide a molecular basis for fine-tuning synaptic function, and open therapeutic directions targeting glycan-binding motifs critical for brain development.


Asunto(s)
Heparitina Sulfato/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Glicopéptidos/análisis , Heparitina Sulfato/química , Humanos , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso , Moléculas de Adhesión de Célula Nerviosa/antagonistas & inhibidores , Moléculas de Adhesión de Célula Nerviosa/genética , Neuronas/citología , Neuronas/metabolismo , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Alineación de Secuencia
2.
Proc Natl Acad Sci U S A ; 119(36): e2112870119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36037373

RESUMEN

Pannexin-1 (Panx1) is a large-pore ion and solute permeable channel highly expressed in the nervous system, where it subserves diverse processes, including neurite outgrowth, dendritic spine formation, and N-methyl D-aspartate (NMDA) receptor (NMDAR)-dependent plasticity. Moreover, Panx1 dysregulation contributes to neurological disorders, including neuropathic pain, epilepsy, and excitotoxicity. Despite progress in understanding physiological and pathological functions of Panx1, the mechanisms that regulate its activity, including its ion and solute permeability, remain poorly understood. In this study, we identify endoplasmic reticulum (ER)-resident stromal interaction molecules (STIM1/2), which are Ca2+ sensors that communicate events within the ER to plasma membrane channels, as binding and signaling partners of Panx1. We demonstrate that Panx1 is activated to its large-pore configuration in response to stimuli that recruit STIM1/2 and map the interaction interface to a hydrophobic region within the N terminus of Panx1. We further characterize a Panx1 N terminus-recognizing antibody as a function-blocking tool able to prevent large-pore Panx1 activation by STIM1/2. Using either the function-blocking antibody or re-expression of Panx1 deletion mutants in Panx1 knockout (KO) neurons, we show that STIM recruitment couples Ca2+ entry via NMDARs to Panx1 activation, thereby identifying a model of NMDAR-STIM-Panx1 signaling in neurons. Our study highlights a previously unrecognized and important role of the Panx1 N terminus in regulating channel activation and membrane localization. Considering past work demonstrating an intimate functional relation between NMDARs and Panx1, our study opens avenues for understanding activation modality and context-specific functions of Panx1, including functions linked to diverse STIM-regulated cellular responses.


Asunto(s)
Calcio , Conexinas , Retículo Endoplásmico , Proteínas del Tejido Nervioso , Receptores de N-Metil-D-Aspartato , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2 , Calcio/metabolismo , Línea Celular , Conexinas/genética , Conexinas/metabolismo , Retículo Endoplásmico/metabolismo , Técnicas de Inactivación de Genes , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo
3.
Mol Psychiatry ; 26(11): 6912-6925, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33981006

RESUMEN

Reduced activity of the mediodorsal thalamus (MD) and abnormal functional connectivity of the MD with the prefrontal cortex (PFC) cause cognitive deficits in schizophrenia. However, the molecular basis of MD hypofunction in schizophrenia is not known. Here, we identified leucine-rich-repeat transmembrane neuronal protein 1 (LRRTM1), a postsynaptic cell-adhesion molecule, as a key regulator of excitatory synaptic function and excitation-inhibition balance in the MD. LRRTM1 is strongly associated with schizophrenia and is highly expressed in the thalamus. Conditional deletion of Lrrtm1 in the MD in adult mice reduced excitatory synaptic function and caused a parallel reduction in the afferent synaptic activity of the PFC, which was reversed by the reintroduction of LRRTM1 in the MD. Our results indicate that chronic reduction of synaptic strength in the MD by targeted deletion of Lrrtm1 functionally disengages the MD from the PFC and may account for cognitive, social, and sensorimotor gating deficits, reminiscent of schizophrenia.


Asunto(s)
Esquizofrenia , Animales , Cognición/fisiología , Proteínas de la Membrana , Ratones , Proteínas del Tejido Nervioso/genética , Corteza Prefrontal , Esquizofrenia/genética , Filtrado Sensorial , Tálamo
4.
J Neurosci ; 40(17): 3348-3359, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32169968

RESUMEN

Nitric oxide (NO) is an important signaling molecule that fulfills diverse functional roles as a neurotransmitter or diffusible second messenger in the developing and adult CNS. Although the impact of NO on different behaviors such as movement, sleep, learning, and memory has been well documented, the identity of its molecular and cellular targets is still an area of ongoing investigation. Here, we identify a novel role for NO in strengthening inhibitory GABAA receptor-mediated transmission in molecular layer interneurons of the mouse cerebellum. NO levels are elevated by the activity of neuronal NO synthase (nNOS) following Ca2+ entry through extrasynaptic NMDA-type ionotropic glutamate receptors (NMDARs). NO activates protein kinase G with the subsequent production of cGMP, which prompts the stimulation of NADPH oxidase and protein kinase C (PKC). The activation of PKC promotes the selective strengthening of α3-containing GABAARs synapses through a GΑΒΑ receptor-associated protein-dependent mechanism. Given the widespread but cell type-specific expression of the NMDAR/nNOS complex in the mammalian brain, our data suggest that NMDARs may uniquely strengthen inhibitory GABAergic transmission in these cells through a novel NO-mediated pathway.SIGNIFICANCE STATEMENT Long-term changes in the efficacy of GABAergic transmission is mediated by multiple presynaptic and postsynaptic mechanisms. A prominent pathway involves crosstalk between excitatory and inhibitory synapses whereby Ca2+-entering through postsynaptic NMDARs promotes the recruitment and strengthening of GABAA receptor synapses via Ca2+/calmodulin-dependent protein kinase II. Although Ca2+ transport by NMDARs is also tightly coupled to nNOS activity and NO production, it has yet to be determined whether this pathway affects inhibitory synapses. Here, we show that activation of NMDARs trigger a NO-dependent pathway that strengthens inhibitory GABAergic synapses of cerebellar molecular layer interneurons. Given the widespread expression of NMDARs and nNOS in the mammalian brain, we speculate that NO control of GABAergic synapse efficacy may be more widespread than has been appreciated.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Cerebelo/metabolismo , Interneuronas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Animales , Cerebelo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Interneuronas/efectos de los fármacos , Masculino , Ratones , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Técnicas de Placa-Clamp , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
5.
Proc Natl Acad Sci U S A ; 115(23): E5382-E5389, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29784826

RESUMEN

Leucine-rich repeat transmembrane (LRRTM) proteins are synaptic cell adhesion molecules that influence synapse formation and function. They are genetically associated with neuropsychiatric disorders, and via their synaptic actions likely regulate the establishment and function of neural circuits in the mammalian brain. Here, we take advantage of the generation of a LRRTM1 and LRRTM2 double conditional knockout mouse (LRRTM1,2 cKO) to examine the role of LRRTM1,2 at mature excitatory synapses in hippocampal CA1 pyramidal neurons. Genetic deletion of LRRTM1,2 in vivo in CA1 neurons using Cre recombinase-expressing lentiviruses dramatically impaired long-term potentiation (LTP), an impairment that was rescued by simultaneous expression of LRRTM2, but not LRRTM4. Mutation or deletion of the intracellular tail of LRRTM2 did not affect its ability to rescue LTP, while point mutations designed to impair its binding to presynaptic neurexins prevented rescue of LTP. In contrast to previous work using shRNA-mediated knockdown of LRRTM1,2, KO of these proteins at mature synapses also caused a decrease in AMPA receptor-mediated, but not NMDA receptor-mediated, synaptic transmission and had no detectable effect on presynaptic function. Imaging of recombinant photoactivatable AMPA receptor subunit GluA1 in the dendritic spines of cultured neurons revealed that it was less stable in the absence of LRRTM1,2. These results illustrate the advantages of conditional genetic deletion experiments for elucidating the function of endogenous synaptic proteins and suggest that LRRTM1,2 proteins help stabilize synaptic AMPA receptors at mature spines during basal synaptic transmission and LTP.


Asunto(s)
Región CA1 Hipocampal/fisiología , Potenciación a Largo Plazo/fisiología , Moléculas de Adhesión de Célula Nerviosa/deficiencia , Células Piramidales/fisiología , Receptores AMPA/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Espinas Dendríticas/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/metabolismo , Células Piramidales/metabolismo , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
7.
Front Mol Neurosci ; 17: 1371145, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38571813

RESUMEN

The prevailing model behind synapse development and specificity is that a multitude of adhesion molecules engage in transsynaptic interactions to induce pre- and postsynaptic assembly. How these extracellular interactions translate into intracellular signal transduction for synaptic assembly remains unclear. Here, we focus on a synapse organizing complex formed by immunoglobulin superfamily member 21 (IgSF21) and neurexin2α (Nrxn2α) that regulates GABAergic synapse development in the mouse brain. We reveal that the interaction between presynaptic Nrxn2α and postsynaptic IgSF21 is a high-affinity receptor-ligand interaction and identify a binding interface in the IgSF21-Nrxn2α complex. Despite being expressed in both dendritic and somatic regions, IgSF21 preferentially regulates dendritic GABAergic presynaptic differentiation whereas another canonical Nrxn ligand, neuroligin2 (Nlgn2), primarily regulates perisomatic presynaptic differentiation. To explore mechanisms that could underlie this compartment specificity, we targeted multiple signaling pathways pharmacologically while monitoring the synaptogenic activity of IgSF21 and Nlgn2. Interestingly, both IgSF21 and Nlgn2 require c-jun N-terminal kinase (JNK)-mediated signaling, whereas Nlgn2, but not IgSF21, additionally requires CaMKII and Src kinase activity. JNK inhibition diminished de novo presynaptic differentiation without affecting the maintenance of formed synapses. We further found that Nrxn2α knockout brains exhibit altered synaptic JNK activity in a sex-specific fashion, suggesting functional linkage between Nrxns and JNK. Thus, our study elucidates the structural and functional relationship of IgSF21 with Nrxn2α and distinct signaling pathways for IgSF21-Nrxn2α and Nlgn2-Nrxn synaptic organizing complexes in vitro. We therefore propose a revised hypothesis that Nrxns act as molecular hubs to specify synaptic properties not only through their multiple extracellular ligands but also through distinct intracellular signaling pathways of these ligands.

8.
Trends Neurosci ; 46(11): 971-985, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37652840

RESUMEN

Synapse organizing proteins are multifaceted molecules that coordinate the complex processes of brain development and plasticity at the level of individual synapses. Their importance is demonstrated by the major brain disorders that emerge when their function is compromised. The mechanisms whereby the various families of organizers govern synapses are diverse, but converge on the structure, function, and plasticity of synapses. Therefore, synapse organizers regulate how synapses adapt to ongoing activity, a process central for determining the developmental trajectory of the brain and critical to all forms of cognition. Here, we explore how synapse organizers set the conditions for synaptic plasticity and the associated molecular events, which eventually link to behavioral features of neurodevelopmental and neuropsychiatric disorders. We also propose central questions on how synapse organizers influence network function through integrating nanoscale and circuit-level organization of the brain.


Asunto(s)
Plasticidad Neuronal , Sinapsis , Humanos , Sinapsis/fisiología , Plasticidad Neuronal/fisiología , Encéfalo , Cognición
9.
Neuron ; 110(13): 2041-2044, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35797957

RESUMEN

Neuronal wiring is facilitated by diverse synaptic adhesion proteins and their repertoire of alternatively spliced isoforms. In this issue of Neuron, Hauser et al. (2022) uncovered the role of a GPI-anchored neurexin 3 splice variant in inhibitory synapse development and dendritic inhibition.


Asunto(s)
Moléculas de Adhesión de Célula Nerviosa , Sinapsis , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sinapsis/metabolismo
10.
Elife ; 112022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35662394

RESUMEN

LRRTMs are postsynaptic cell adhesion proteins that have region-restricted expression in the brain. To determine their role in the molecular organization of synapses in vivo, we studied synapse development and plasticity in hippocampal neuronal circuits in mice lacking both Lrrtm1 and Lrrtm2. We found that LRRTM1 and LRRTM2 regulate the density and morphological integrity of excitatory synapses on CA1 pyramidal neurons in the developing brain but are not essential for these roles in the mature circuit. Further, they are required for long-term-potentiation in the CA3-CA1 pathway and the dentate gyrus, and for enduring fear memory in both the developing and mature brain. Our data show that LRRTM1 and LRRTM2 regulate synapse development and function in a cell-type and developmental-stage-specific manner, and thereby contribute to the fine-tuning of hippocampal circuit connectivity and plasticity.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa , Animales , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Ratones , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Sinapsis/fisiología
11.
J Neurosci ; 30(22): 7495-506, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20519524

RESUMEN

Leucine-rich repeat transmembrane neuronal proteins (LRRTMs) were recently found to instruct presynaptic and mediate postsynaptic glutamatergic differentiation. In a candidate screen, here we identify neurexin-1beta lacking an insert at splice site 4 (-S4) as a ligand for LRRTM2. Neurexins bind LRRTM2 with a similar affinity but distinct code from the code for binding neuroligin-1 (the predominant form of neuroligin-1 at glutamate synapses, containing the B splice site insert). Whereas neuroligin-1 binds to neurexins 1, 2, and 3 beta but not alpha variants, regardless of insert at splice site 4, LRRTM2 binds to neurexins 1, 2, and 3 alpha and beta variants specifically lacking an insert at splice site 4. We further show that this binding code is conserved in LRRTM1, the family member linked to schizophrenia and handedness, and that the code is functional in a coculture hemisynapse formation assay. Mutagenesis of LRRTM2 to prevent binding to neurexins abolishes presynaptic inducing activity of LRRTM2. Remarkably, mutagenesis of neurexins shows that the binding face on neurexin-1beta (-S4) is highly overlapping for the structurally distinct LRRTM2 and neuroligin-1 partners. Finally, we explore here the interplay of neuroligin-1 and LRRTM2 in synapse regulation. In neuron cultures, LRRTM2 is more potent than neuroligin-1 in promoting synaptic differentiation, and, most importantly, these two families of neurexin-binding partners cooperate in an additive or synergistic manner. Thus, we propose a synaptic code hypothesis suggesting that neurexins are master regulators of the cooperative activities of LRRTMs and neuroligins.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Ácido Glutámico/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/fisiología , Sinapsis/fisiología , Empalme Alternativo/genética , Animales , Calcio/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Diferenciación Celular , Células Cultivadas , Chlorocebus aethiops , Propuestas de Licitación/métodos , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Humanos , Mutagénesis/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Transfección/métodos
12.
Hum Genet ; 130(4): 563-73, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21424692

RESUMEN

Growing genetic evidence is converging in favor of common pathogenic mechanisms for autism spectrum disorders (ASD), intellectual disability (ID or mental retardation) and schizophrenia (SCZ), three neurodevelopmental disorders affecting cognition and behavior. Copy number variations and deleterious mutations in synaptic organizing proteins including NRXN1 have been associated with these neurodevelopmental disorders, but no such associations have been reported for NRXN2 or NRXN3. From resequencing the three neurexin genes in individuals affected by ASD (n = 142), SCZ (n = 143) or non-syndromic ID (n = 94), we identified a truncating mutation in NRXN2 in a patient with ASD inherited from a father with severe language delay and family history of SCZ. We also identified a de novo truncating mutation in NRXN1 in a patient with SCZ, and other potential pathogenic ASD mutations. These truncating mutations result in proteins that fail to promote synaptic differentiation in neuron coculture and fail to bind either of the established postsynaptic binding partners LRRTM2 or NLGN2 in cell binding assays. Our findings link NRXN2 disruption to the pathogenesis of ASD for the first time and further strengthen the involvement of NRXN1 in SCZ, supporting the notion of a common genetic mechanism in these disorders.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Trastornos Generalizados del Desarrollo Infantil/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Esquizofrenia/genética , Secuencia de Aminoácidos , Animales , Células COS , Proteínas de Unión al Calcio , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Cultivadas , Niño , Chlorocebus aethiops , Estudios de Cohortes , Femenino , Dosificación de Gen , Predisposición Genética a la Enfermedad , Humanos , Trastornos del Desarrollo del Lenguaje/genética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa , Neuronas/citología , Neuronas/metabolismo , Linaje , Homología de Secuencia de Aminoácido
13.
Neuron ; 109(8): 1248-1250, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33887189

RESUMEN

Competition between active and inactive synapses sculpts neuronal networks by activity-dependent loss of inactive connections, the mechanisms for which are poorly understood. In this issue of Neuron, Yasuda et al. (2021) demonstrate that JAK2-STAT1 signaling in inactive axons and synapses is essential for their elimination.


Asunto(s)
Axones , Sinapsis , Animales , Encéfalo , Neuronas
14.
Psychoneuroendocrinology ; 126: 105147, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33497916

RESUMEN

Obesity before and during pregnancy negatively affects the mental and physical health of the mother. A diet high in fat also increases the risk for anxiety, depression and cognitive dysfunction. We examined the effects of high fat diet (HFD) -induced obesity and pregnancy on maternal behavior, cognitive function and anxiety- and depression-like behaviors in mice. Four-week-old female CD-1 mice were placed on a HFD or regular chow diet (RCD) for 5 weeks. Mice were maintained on either diet as non-pregnant HFD and RCD groups, or allowed to breed, and then fed these diets throughout gestation, lactation and after weaning, as pregnant HFD and RCD groups. Mice on HFD but not on RCD for 5 weeks pre-pregnancy significantly gained weight and had impaired glucose clearance. Maternal behavior was assessed by nest building prepartum and pup-retrieval postpartum. Anxiety-like behavior was evaluated both prepartum and postpartum by elevated plus maze and cognitive function was assessed by the novel object recognition test postpartum. Anhedonia, a measure of impaired reward function, is an endophenotype of depression and was assessed by sucrose preference test pre- and post-weaning in dams. Mice on HFD in pregnancy exhibited both impaired maternal behavior and cognitive function in the postpartum period. We did not detect measurable differences between the HFD and RCD groups in anxiety-like behavior in the prepartum period. In contrast, HFD was also associated with anhedonia in pregnant mice pre-weaning, and anxiety-like behavior post-weaning. Thus, HFD has a negative effect on maternal behavior in the outbred CD-1 mouse, which provides a model to study associated outcomes and related mechanisms.


Asunto(s)
Anhedonia , Dieta Alta en Grasa , Conducta Materna , Obesidad , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Lactancia , Ratones , Obesidad/etiología , Embarazo
15.
Mol Biol Cell ; 18(6): 2037-46, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17360966

RESUMEN

Neuronal exocytosis is driven by the formation of SNARE complexes between synaptobrevin 2 on synaptic vesicles and SNAP-25/syntaxin 1 on the plasma membrane. It has remained controversial, however, whether SNAREs are constitutively active or whether they are down-regulated until fusion is triggered. We now show that synaptobrevin in proteoliposomes as well as in purified synaptic vesicles is constitutively active. Potential regulators such as calmodulin or synaptophysin do not affect SNARE activity. Substitution or deletion of residues in the linker connecting the SNARE motif and transmembrane region did not alter the kinetics of SNARE complex assembly or of SNARE-mediated fusion of liposomes. Remarkably, deletion of C-terminal residues of the SNARE motif strongly reduced fusion activity, although the overall stability of the complexes was not affected. We conclude that although complete zippering of the SNARE complex is essential for membrane fusion, the structure of the adjacent linker domain is less critical, suggesting that complete SNARE complex assembly not only connects membranes but also drives fusion.


Asunto(s)
Membrana Celular/metabolismo , Exocitosis/fisiología , Neuronas/fisiología , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Calmodulina/metabolismo , Humanos , Fusión de Membrana/fisiología , Datos de Secuencia Molecular , Neuronas/citología , Proteolípidos/metabolismo , Proteínas R-SNARE/genética , Ratas , Proteínas SNARE/genética , Alineación de Secuencia , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Sinaptofisina/metabolismo
16.
Neuron ; 105(6): 1007-1017.e5, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31974009

RESUMEN

LRRTM4 is a transsynaptic adhesion protein regulating glutamatergic synapse assembly on dendrites of central neurons. In the mouse retina, we find that LRRTM4 is enriched at GABAergic synapses on axon terminals of rod bipolar cells (RBCs). Knockout of LRRTM4 reduces RBC axonal GABAA and GABAC receptor clustering and disrupts presynaptic inhibition onto RBC terminals. LRRTM4 removal also perturbs the stereotyped output synapse arrangement at RBC terminals. Synaptic ribbons are normally apposed to two distinct postsynaptic "dyad" partners, but in the absence of LRRTM4, "monad" and "triad" arrangements are also formed. RBCs from retinas deficient in GABA release also demonstrate dyad mis-arrangements but maintain LRRTM4 expression, suggesting that defects in dyad organization in the LRRTM4 knockout could originate from reduced GABA receptor function. LRRTM4 is thus a key synapse organizing molecule at RBC terminals, where it regulates function of GABAergic synapses and assembly of RBC synaptic dyads.


Asunto(s)
Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Inhibición Neural/fisiología , Terminales Presinápticos/fisiología , Células Bipolares de la Retina/fisiología , Animales , Femenino , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Receptores de GABA/metabolismo , Receptores de GABA/fisiología , Retina/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Ácido gamma-Aminobutírico/metabolismo
17.
Neuron ; 106(1): 108-125.e12, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-31995730

RESUMEN

Presynaptic neurexins (Nrxs) and type IIa receptor-type protein tyrosine phosphatases (RPTPs) organize synapses through a network of postsynaptic ligands. We show that leucine-rich-repeat transmembrane neuronal proteins (LRRTMs) differentially engage the protein domains of Nrx but require its heparan sulfate (HS) modification to induce presynaptic differentiation. Binding to the HS of Nrx is sufficient for LRRTM3 and LRRTM4 to induce synaptogenesis. We identify mammalian Nrx1γ as a potent synapse organizer and reveal LRRTM4 as its postsynaptic ligand. Mice expressing a mutant form of LRRTM4 that cannot bind to HS show structural and functional deficits at dentate gyrus excitatory synapses. Through the HS of Nrx, LRRTMs also recruit PTPσ to induce presynaptic differentiation but function to varying degrees in its absence. PTPσ forms a robust complex with Nrx, revealing an unexpected interaction between the two presynaptic hubs. These findings underscore the complex interplay of synapse organizers in specifying the molecular logic of a neural circuit.


Asunto(s)
Proteínas de Unión al Calcio/genética , Giro Dentado/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Sinapsis/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Giro Dentado/patología , Heparitina Sulfato/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Sinapsis/patología
18.
Artículo en Inglés | MEDLINE | ID: mdl-31191292

RESUMEN

Formation of synapses between neurons depends in part on binding between axonal and dendritic cell surface synaptic organizing proteins, which recruit components of the developing presynaptic and postsynaptic specializations. One of these presynaptic organizing molecules is protein tyrosine phosphatase σ (PTPσ). Although the protein domains involved in adhesion between PTPσ and its postsynaptic binding partners are known, the mechanisms by which it signals into the presynaptic neuron to recruit synaptic vesicles and other necessary components for regulated transmitter release are not well understood. One attractive candidate to mediate this function is liprin-α, a scaffolding protein with well-established roles at the synapse. We systematically mutated residues of the PTPσ intracellular region (ICR) and used the yeast dihydrofolate reductase (DHFR) protein complementation assay to screen for disrupted interactions between these mutant forms of PTPσ and its various binding partners. Using a molecular replacement strategy, we show that disrupting the interaction between PTPσ and liprin-α, but not between PTPσ and itself or another binding partner, caskin, abolishes presynaptic differentiation. Furthermore, phosphatase activity of PTPσ and binding to extracellular heparan sulfate (HS) proteoglycans are dispensable for presynaptic induction. Previous reports have suggested that binding between PTPσ and liprin-α is mediated by the PTPσ membrane-distal phosphatase-like domain. However, we provide evidence here that both of the PTPσ phosphatase-like domains mediate binding to liprin-α and are required for PTPσ-mediated presynaptic differentiation. These findings further our understanding of the mechanistic basis by which PTPσ acts as a presynaptic organizer.

19.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3353-3367, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30055293

RESUMEN

Cardiolipin (CL) is a key mitochondrial phospholipid essential for mitochondrial energy production. CL is remodeled from monolysocardiolipin (MLCL) by the enzyme tafazzin (TAZ). Loss-of-function mutations in the gene which encodes TAZ results in a rare X-linked disorder called Barth Syndrome (BTHS). The mutated TAZ is unable to maintain the physiological CL:MLCL ratio, thus reducing CL levels and affecting mitochondrial function. BTHS is best known as a cardiac disease, but has been acknowledged as a multi-syndrome disorder, including cognitive deficits. Since reduced CL levels has also been reported in numerous neurodegenerative disorders, we examined how TAZ-deficiency impacts cognitive abilities, brain mitochondrial respiration and the function of hippocampal neurons and glia in TAZ knockdown (TAZ kd) mice. We have identified for the first time the profile of changes that occur in brain phospholipid content and composition of TAZ kd mice. The brain of TAZ kd mice exhibited reduced TAZ protein expression, reduced total CL levels and a 19-fold accumulation of MLCL compared to wild-type littermate controls. TAZ kd brain exhibited a markedly distinct profile of CL and MLCL molecular species. In mitochondria, the activity of complex I was significantly elevated in the monomeric and supercomplex forms with TAZ-deficiency. This corresponded with elevated mitochondrial state I respiration and attenuated spare capacity. Furthermore, the production of reactive oxygen species was significantly elevated in TAZ kd brain mitochondria. While motor function remained normal in TAZ kd mice, they showed significant memory deficiency based on novel object recognition test. These results correlated with reduced synaptophysin protein levels and derangement of the neuronal CA1 layer in hippocampus. Finally, TAZ kd mice had elevated activation of brain immune cells, microglia compared to littermate controls. Collectively, our findings demonstrate that TAZ-mediated remodeling of CL contributes significantly to the expansive distribution of CL molecular species in the brain, plays a key role in mitochondria respiratory activity, maintains normal cognitive function, and identifies the hippocampus as a potential therapeutic target for BTHS.


Asunto(s)
Cardiolipinas/metabolismo , Disfunción Cognitiva/genética , Hipocampo/metabolismo , Factores de Transcripción/genética , Aciltransferasas , Animales , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Lisofosfolípidos , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sinaptofisina/metabolismo , Factores de Transcripción/metabolismo
20.
Methods Mol Biol ; 1538: 83-91, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27943185

RESUMEN

This protocol describes an in situ protein-protein interaction assay between tagged recombinant proteins and cell-surface expressed synaptic proteins. The assay is arguably more sensitive than other traditional protein binding assays such as co-immunoprecipitation and pull-downs and provides a visual readout for binding. This assay has been widely used to determine the dissociation constant of binding of trans-synaptic adhesion proteins. The step-wise description in the protocol should facilitate the adoption of this method in other laboratories.


Asunto(s)
Proteínas Portadoras/metabolismo , Fragmentos Fc de Inmunoglobulinas/genética , Mapeo de Interacción de Proteínas/métodos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Animales , Células COS , Técnicas de Cultivo de Célula , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Expresión Génica , Células HEK293 , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Microscopía Fluorescente , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA