Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Neurosci ; 123: 103783, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36208859

RESUMEN

Cytosolic PSD-95 interactor (cypin) is a multifunctional, guanine deaminase that plays a major role in shaping the morphology of the dendritic arbor of hippocampal and cortical neurons. Cypin catalyzes the Zn2+-dependent deamination of guanine to xanthine, which is then metabolized to uric acid by xanthine oxidase. Cypin binds to tubulin heterodimers via its carboxyl terminal region (amino acids (aa) 350-454), which contains a collapsin response mediator protein (CRMP) homology domain (aa 350-403). Moreover, this region alone is not sufficient to facilitate microtubule polymerization; therefore, additional cypin regions must be involved in this process. Here, we asked whether cypin binds to fully formed microtubules and how overexpression of cypin regulates the microtubule cytoskeleton in dendrites of cultured hippocampal neurons. Protein-protein docking strategies confirm that the cypin homodimer binds to tubulin heterodimers via amino acids within aa 350-454. Biochemical pull-down data suggest that aa 1-220 are necessary for cypin binding to soluble tubulin heterodimers and to taxol-stabilized microtubules. Molecular docking of the cypin homodimer to soluble tubulin heterodimers reveals a consistently observed docking pose using aa 47-71, 113-118, 174-178, and 411-418, which is consistent with our biochemical data. Additionally, overexpression of cypin in hippocampal neurons results in decreased spacing between microtubules. Our results suggest that several protein domains facilitate cypin-mediated polymerization of tubulin heterodimers into microtubules, possibly through a mechanism whereby cypin dimers bind to multiple tubulin heterodimers.


Asunto(s)
Dendritas , Tubulina (Proteína) , Tubulina (Proteína)/metabolismo , Dendritas/metabolismo , Simulación del Acoplamiento Molecular , Proteínas Portadoras/metabolismo , Neuronas/metabolismo , Hipocampo/metabolismo , Microtúbulos/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Aminoácidos/metabolismo
2.
Mol Cell Neurosci ; 109: 103562, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32987141

RESUMEN

Abnormal dendritic arbor development has been implicated in a number of neurodevelopmental disorders, such as autism and Rett syndrome, and the neuropsychiatric disorder schizophrenia. Postmortem brain samples from subjects with schizophrenia show elevated levels of NOS1AP in the dorsolateral prefrontal cortex, a region of the brain associated with cognitive function. We previously reported that the long isoform of NOS1AP (NOS1AP-L), but not the short isoform (NOS1AP-S), negatively regulates dendrite branching in rat hippocampal neurons. To investigate the role that NOS1AP isoforms play in human dendritic arbor development, we adapted methods to generate human neural progenitor cells and neurons using induced pluripotent stem cell (iPSC) technology. We found that increased protein levels of either NOS1AP-L or NOS1AP-S decrease dendrite branching in human neurons at the developmental time point when primary and secondary branching actively occurs. Next, we tested whether pharmacological agents can decrease the expression of NOS1AP isoforms. Treatment of human iPSC-derived neurons with d-serine, but not clozapine, haloperidol, fluphenazine, or GLYX-13, results in a reduction in endogenous NOS1AP-L, but not NOS1AP-S, protein expression; however, d-serine treatment does not reverse decreases in dendrite number mediated by overexpression of NOS1AP isoforms. In summary, we demonstrate how an in vitro model of human neuronal development can help in understanding the etiology of schizophrenia and can also be used as a platform to screen drugs for patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Dendritas/ultraestructura , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Neuronas/citología , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Células Cultivadas , Clozapina/farmacología , Evaluación Preclínica de Medicamentos , Flufenazina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/fisiología , Haloperidol/farmacología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Canales Iónicos/fisiología , Proteínas del Tejido Nervioso/fisiología , Células-Madre Neurales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oligopéptidos/farmacología , Técnicas de Placa-Clamp , Isoformas de Proteínas/fisiología , Esquizofrenia/etiología , Esquizofrenia/genética , Serina/farmacología
3.
Cereb Cortex ; 29(7): 2890-2903, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29982499

RESUMEN

Higher brain function relies on proper development of the cerebral cortex, including correct positioning of neurons and dendrite morphology. Disruptions in these processes may result in various neurocognitive disorders. Mutations in the CPE gene, which encodes carboxypeptidase E (CPE), have been linked to depression and intellectual disability. However, it remains unclear whether CPE is involved in early brain development and in turn contributes to the pathophysiology of neurocognitive disorders. Here, we investigate the effects of CPE knockdown on early brain development and explore the functional significance of the interaction between CPE and its binding partner p150Glued. We demonstrate that CPE is required for cortical neuron migration and dendrite arborization. Furthermore, we show that expression of CPE-C10 redistributes p150Glued from the centrosome and that disruption of CPE interaction with p150Glued leads to abnormal neuronal migration and dendrite morphology, suggesting that a complex between CPE and p150Glued is necessary for proper neurodevelopment.


Asunto(s)
Carboxipeptidasa H/metabolismo , Corteza Cerebral/fisiología , Dendritas/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Animales , Células COS , Movimiento Celular/fisiología , Corteza Cerebral/embriología , Chlorocebus aethiops , Ratones , Ratas
4.
J Biol Chem ; 293(5): 1568-1569, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29414768

RESUMEN

Long-term depression (LTD) is a reduction in the efficacy of neuronal synapses, but the molecular basis of LTD signaling and how these signals lead to phenotypic outcomes, such as the shrinkage of synaptic regions, is not clear. In a new report, Woolfrey et al use chemically-induced LTD and a multitude of in vitro biochemical assays to provide evidence that synaptic removal of the scaffolding protein AKAP79/150 promotes LTD-induced spine shrinkage. The further identification of CaMKII, a kinase primarily associated with long-term potentiation (LTP), as a requirement for AKAP79/150 removal, uncovers unexpected interplay between different post-translational modifications and points to a new model of LTD.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Depresión Sináptica a Largo Plazo , Fosfolípidos/metabolismo , Membranas Sinápticas/metabolismo , Animales , Lipoilación , Fosforilación , Dominios Proteicos , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Columna Vertebral/metabolismo , Columna Vertebral/patología , Membranas Sinápticas/patología
6.
Mol Cell Neurosci ; 89: 20-32, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29601869

RESUMEN

Antipsychotic medications are inefficient at treating symptoms of schizophrenia (SCZ), and N-methyl d-aspartate receptor (NMDAR) agonists are potential therapeutic alternatives. As such, these agonists may act on different pathways and proteins altered in the brains of patients with SCZ than do antipsychotic medications. Here, we investigate the effects of administration of the antipsychotic haloperidol and NMDAR agonist d-serine on function and expression of three proteins that play significant roles in SCZ: nitric oxide synthase 1 adaptor protein (NOS1AP), dopamine D2 (D2) receptor, and disrupted in schizophrenia 1 (DISC1). We administered haloperidol or d-serine to male and female Sprague Dawley rats via intraperitoneal injection for 12 days and subsequently examined cortical expression of NOS1AP, D2 receptor, and DISC1. We found sex-specific effects of haloperidol and d-serine treatment on the expression of these proteins. Haloperidol significantly reduced expression of D2 receptor in male, but not female, rats. Conversely, d-serine reduced expression of NOS1AP in male rats and did not affect D2 receptor expression. d-serine treatment also reduced expression of DISC1 in male rats and increased DISC1 expression in female rats. As NOS1AP is overexpressed in the cortex of patients with SCZ and negatively regulates NMDAR signaling, we subsequently examined whether treatment with antipsychotics or NMDAR agonists can reverse the detrimental effects of NOS1AP overexpression in vitro as previously reported by our group. NOS1AP overexpression promotes reduced dendrite branching in vitro, and as such, we treated cortical neurons overexpressing NOS1AP with different antipsychotics (haloperidol, clozapine, fluphenazine) or d-serine for 24 h and determined the effects of these drugs on NOS1AP expression and dendrite branching. While antipsychotics did not affect NOS1AP protein expression or dendrite branching in vitro, d-serine reduced NOS1AP expression and rescued NOS1AP-mediated reductions in dendrite branching. Taken together, our data suggest that d-serine influences the function and expression of NOS1AP, D2 receptor, and DISC1 in a sex-specific manner and reverses the effects of NOS1AP overexpression on dendrite morphology.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Corteza Cerebral/efectos de los fármacos , Neuronas/efectos de los fármacos , Serina/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antipsicóticos/farmacología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Femenino , Haloperidol/farmacología , Isomerismo , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Serina/química , Factores Sexuales
7.
Neurobiol Dis ; 119: 13-25, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30031156

RESUMEN

Cytosolic PSD-95 interactor (cypin), the primary guanine deaminase in the brain, plays key roles in shaping neuronal circuits and regulating neuronal survival. Despite this pervasive role in neuronal function, the ability for cypin activity to affect recovery from acute brain injury is unknown. A key barrier in identifying the role of cypin in neurological recovery is the absence of pharmacological tools to manipulate cypin activity in vivo. Here, we use a small molecule screen to identify two activators and one inhibitor of cypin's guanine deaminase activity. The primary screen identified compounds that change the initial rate of guanine deamination using a colorimetric assay, and secondary screens included the ability of the compounds to protect neurons from NMDA-induced injury and NMDA-induced decreases in frequency and amplitude of miniature excitatory postsynaptic currents. Hippocampal neurons pretreated with activators preserved electrophysiological function and survival after NMDA-induced injury in vitro, while pretreatment with the inhibitor did not. The effects of the activators were abolished when cypin was knocked down. Administering either cypin activator directly into the brain one hour after traumatic brain injury significantly reduced fear conditioning deficits 5 days after injury, while delivering the cypin inhibitor did not improve outcome after TBI. Together, these data demonstrate that cypin activation is a novel approach for improving outcome after TBI and may provide a new pathway for reducing the deficits associated with TBI in patients.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/prevención & control , Guanina Desaminasa/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/fisiopatología , Células COS , Células Cultivadas , Chlorocebus aethiops , Dimetilsulfóxido/farmacología , Miedo/efectos de los fármacos , Miedo/fisiología , Guanina Desaminasa/antagonistas & inhibidores , Compuestos Heterocíclicos con 3 Anillos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato/farmacología , Técnicas de Cultivo de Órganos , Ratas
8.
Biotechnol Bioeng ; 115(4): 815-830, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29251352

RESUMEN

Traumatic brain injury (TBI) affects 5.3 million people in the United States, and there are 12,500 new cases of spinal cord injury (SCI) every year. There is yet a significant need for in vitro models of TBI and SCI in order to understand the biological mechanisms underlying central nervous system (CNS) injury and to identify and test therapeutics to aid in recovery from neuronal injuries. While TBI or SCI studies have been aided with traditional in vivo and in vitro models, the innate limitations in specificity of injury, isolation of neuronal regions, and reproducibility of these models can decrease their usefulness in examining the neurobiology of injury. Microfluidic devices provide several advantages over traditional methods by allowing researchers to (1) examine the effect of injury on specific neural components, (2) fluidically isolate neuronal regions to examine specific effects on subcellular components, and (3) reproducibly create a variety of injuries to model TBI and SCI. These microfluidic devices are adaptable for modeling a wide range of injuries, and in this review, we will examine different methodologies and models recently utilized to examine neuronal injury. Specifically, we will examine vacuum-assisted axotomy, physical injury, chemical injury, and laser-based axotomy. Finally, we will discuss the benefits and downsides to each type of injury model and discuss how researchers can use these parameters to pick a particular microfluidic device to model CNS injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Técnicas In Vitro , Dispositivos Laboratorio en un Chip , Traumatismos de la Médula Espinal/metabolismo , Animales , Axotomía , Humanos , Neuronas/patología , Vacio
9.
Cell Mol Life Sci ; 74(23): 4369-4385, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28698933

RESUMEN

Proper communication among neurons depends on an appropriately formed dendritic arbor, and thus, aberrant changes to the arbor are implicated in many pathologies, ranging from cognitive disorders to neurodegenerative diseases. Due to the importance of dendritic shape to neuronal network function, the morphology of dendrites is tightly controlled and is influenced by both intrinsic and extrinsic factors. In this work, we examine how brain-derived neurotrophic factor (BDNF), one of the most well-studied extrinsic regulators of dendritic branching, affects the arbor when it is applied locally via microbeads to cultures of hippocampal neurons. We found that local application of BDNF increases both proximal and distal branching in a time-dependent manner and that local BDNF application attenuates pruning of dendrites that occurs with neuronal maturation. Additionally, we examined whether cytosolic PSD-95 interactor (cypin), an intrinsic regulator of dendritic branching, plays a role in these changes and found strong evidence for the involvement of cypin in BDNF-promoted increases in dendrites after 24 but not 48 h of application. This current study extends our previous work in which we found that bath application of BDNF for 72 h, but not shorter times, increases proximal dendrite branching and that this increase occurs through transcriptional regulation of cypin. Moreover, this current work illustrates how dendritic branching is regulated differently by the same growth factor depending on its spatial localization, suggesting a novel pathway for modulation of dendritic branching locally.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Dendritas/efectos de los fármacos , Hipocampo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Plasticidad Neuronal/efectos de los fármacos , Animales , Dendritas/metabolismo , Dendritas/ultraestructura , Homólogo 4 de la Proteína Discs Large , Embrión de Mamíferos , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Hipocampo/ultraestructura , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Microesferas , Plasticidad Neuronal/genética , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Factores de Tiempo , Transcripción Genética , Transfección
10.
Hum Genet ; 136(1): 67-74, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27726012

RESUMEN

More than 31,000 protein-coding sequences (CCDS) have been identified in the human genome. Here, we analyzed codon usage in all human CCDS and found that there is a preferential usage of minor codons for Ala (CGC), Pro (CCG), Ser (UCG), and Thr (ACG) in the initial 50-codon sequences of the CCDS. These codons, with consensus XCG sequences, are most infrequently used among their synonymous codons. Thus, the tRNA concentrations per codon are considered to be highest for the minor codons for Ala, Pro, Ser and Thr in comparison with other synonymous codons for each of them to enhance the translation efficiency. This suggests that human genes are regulated at the level of translation by preferentially using minor codons within the first 50 codons of the CCDS. This hypothesis was experimentally confirmed by comparing the expression of the luciferase gene encoded by minor codons with that encoded by major codons.


Asunto(s)
Codón , Genoma Humano , Iniciación de la Cadena Peptídica Traduccional/genética , Regulación de la Expresión Génica , Humanos , Sistemas de Lectura Abierta
11.
J Cell Physiol ; 229(9): 1224-35, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24435709

RESUMEN

Neurons have a limited capacity for heat shock protein (HSP) induction and are vulnerable to the pathogenic consequence of protein misfolding and aggregation as seen in age-related neurodegenerative diseases. Sirtuin 1 (SIRT1), an NAD(+) -dependent lysine deacetylase with important biological functions, has been shown to sustain the DNA-binding state of HSF1 for HSP induction. Here we show that differentiation and maturation of embryonic cortical neurons and N2a neuroprogenitor cells is associated with decreases in SIRT1 expression and heat shock-dependent induction of HSP70 protein. Tests of a pharmacological activator and an inhibitor of SIRT1 affirm the regulatory role of SIRT1 in HSP70 induction. Protein cross-linking studies show that nuclear SIRT1 and HSF1 form a co-migrating high molecular weight complex upon stress. The use of retroviral vectors to manipulate SIRT1 expression in N2a cells show that shRNA-mediated knock down of SIRT1 causes spontaneous neurite outgrowth coincident with reduced growth rate and decreased induction of hsp70-reporter gene, whereas SIRT1 over-expression blocks the induced neural differentiation of N2a cells. Our results suggest that decreased SIRT1 expression is conducive to neuronal differentiation and this decrease contributes to the attenuated induction of HSPs in neurons.


Asunto(s)
Corteza Cerebral/enzimología , Técnicas de Silenciamiento del Gen , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico , Células-Madre Neurales/enzimología , Neurogénesis , Neuronas/enzimología , Sirtuina 1/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Corteza Cerebral/embriología , Corteza Cerebral/patología , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Edad Gestacional , Proteínas HSP70 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/genética , Ratones , Células-Madre Neurales/patología , Neuronas/patología , Unión Proteica , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Sirtuina 1/genética , Factores de Tiempo , Factores de Transcripción/metabolismo , Transfección
12.
Brain ; 136(Pt 1): 65-80, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23293266

RESUMEN

The recent public awareness of the incidence and possible long-term consequences of traumatic brain injury only heightens the need to develop effective approaches for treating this neurological disease. In this report, we identify a new therapeutic target for traumatic brain injury by studying the role of astrocytes, rather than neurons, after neurotrauma. We use in vivo multiphoton imaging and show that mechanical forces during trauma trigger intercellular calcium waves throughout the astrocytes, and these waves are mediated by purinergic signalling. Subsequent in vitro screening shows that astrocyte signalling through the 'mechanical penumbra' affects the activity of neural circuits distant from the injury epicentre, and a reduction in the intercellular calcium waves within astrocytes restores neural activity after injury. In turn, the targeting of different purinergic receptor populations leads to a reduction in hippocampal cell death in mechanically injured organotypic slice cultures. Finally, the most promising therapeutic candidate from our in vitro screen (MRS 2179, a P2Y1 receptor antagonist) also improves histological and cognitive outcomes in a preclinical model of traumatic brain injury. This work shows the potential of studying astrocyte signalling after trauma to yield new and effective therapeutic targets for treating traumatic brain injury.


Asunto(s)
Adenosina Difosfato/análogos & derivados , Astrocitos/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2Y/farmacología , Recuperación de la Función/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Adenosina Difosfato/farmacología , Adenosina Difosfato/uso terapéutico , Animales , Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/lesiones , Corteza Cerebral/metabolismo , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Ratas , Ratas Sprague-Dawley
13.
Mol Neurobiol ; 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38589756

RESUMEN

α-Klotho (α-Kl) is a modulator of aging, neuroprotection, and cognition. Transcription of the Klotho gene produces two splice variants-a membrane protein (mKl), which can be cleaved and released into the extracellular milieu, and a truncated secreted form (sKl). Despite mounting evidence supporting a role for α-Kl in brain function, the specific roles of α-Kl isoforms in neuronal development remain elusive. Here, we examined α-Kl protein levels in rat brain and observed region-specific expression in the adult that differs between isoforms. In the developing hippocampus, levels of isoforms decrease after the third postnatal week, marking the end of the critical period for development. We overexpressed α-Kl isoforms in primary cultures of rat cortical neurons and evaluated effects on brain-derived neurotrophic factor (BDNF) signaling. Overexpression of either isoform attenuated BDNF-mediated signaling and reduced intracellular Ca2+ levels, with mKl promoting a greater effect. mKl or sKl overexpression in hippocampal neurons resulted in a partially overlapping reduction in secondary dendrite branching. Moreover, mKl overexpression increased primary dendrite number. BDNF treatment of neurons overexpressing sKl resulted in a dendrite branching phenotype similar to control neurons. In neurons overexpressing mKl, BDNF treatment restored branching of secondary and higher order dendrites close, but not distal, to the soma. Taken together, the data presented support the idea that sKl and mKl play distinct roles in neuronal development, and specifically, in dendrite morphogenesis.

14.
bioRxiv ; 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38712188

RESUMEN

While recent work has identified roles for immune mediators in the regulation of neural activity, the capacity for cell intrinsic innate immune signaling within neurons to influence neurotransmission remains poorly understood. However, the existing evidence linking immune signaling with neuronal function suggests that modulation of neurotransmission may serve previously undefined roles in host protection during infection of the central nervous system. Here, we identify a specialized function for RIPK3, a kinase traditionally associated with necroptotic cell death, in preserving neuronal survival during neurotropic flavivirus infection through the suppression of excitatory neurotransmission. We show that RIPK3 coordinates transcriptomic changes in neurons that suppress neuronal glutamate signaling, thereby desensitizing neurons to excitotoxic cell death. These effects occur independently of the traditional functions of RIPK3 in promoting necroptosis and inflammatory transcription. Instead, RIPK3 promotes phosphorylation of the key neuronal regulatory kinase CaMKII, which in turn activates the transcription factor CREB to drive a neuroprotective transcriptional program and suppress deleterious glutamatergic signaling. These findings identify an unexpected function for a canonical cell death protein in promoting neuronal survival during viral infection through the modulation of neuronal activity, highlighting new mechanisms of neuroimmune crosstalk.

15.
eNeuro ; 11(2)2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38302457

RESUMEN

Cypin (cytosolic postsynaptic density protein 95 interactor) is the primary guanine deaminase in the central nervous system (CNS), promoting the metabolism of guanine to xanthine, an important reaction in the purine salvage pathway. Activation of the purine salvage pathway leads to the production of uric acid (UA). UA has paradoxical effects, specifically in the context of CNS injury as it confers neuroprotection, but it also promotes pain. Since neuropathic pain is a comorbidity associated with spinal cord injury (SCI), we postulated that small molecule cypin inhibitor B9 treatment could attenuate SCI-induced neuropathic pain, potentially by interfering with UA production. However, we also considered that this treatment could hinder the neuroprotective effects of UA and, in doing so, exacerbate SCI outcomes. To address our hypothesis, we induced a moderate midthoracic contusion SCI in female mice and assessed whether transient intrathecal administration of B9, starting at 1 d postinjury (dpi) until 7 dpi, attenuates mechanical pain in hindlimbs at 3 weeks pi. We also evaluated the effects of B9 on the spontaneous recovery of locomotor function. We found that B9 alleviates mechanical pain but does not affect locomotor function. Importantly, B9 does not exacerbate lesion volume at the epicenter. In accordance with these findings, B9 does not aggravate glutamate-induced excitotoxic death of SC neurons in vitro. Moreover, SCI-induced increased astrocyte reactivity at the glial scar is not altered by B9 treatment. Our data suggest that B9 treatment reduces mechanical pain without exerting major detrimental effects following SCI.


Asunto(s)
Neuralgia , Traumatismos de la Médula Espinal , Ratones , Femenino , Animales , Hiperalgesia/metabolismo , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Neuronas/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Neuralgia/metabolismo , Purinas , Médula Espinal/metabolismo
16.
Ann Biomed Eng ; 52(4): 1021-1038, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38294641

RESUMEN

Traumatic brain injury (TBI) is the leading cause of accident-related death and disability in the world and can lead to long-term neuropsychiatric symptoms, such as a decline in cognitive function and neurodegeneration. TBI includes primary and secondary injury, with head trauma and deformation of the brain caused by the physical force of the impact as primary injury, and cellular and molecular cascades that lead to cell death as secondary injury. Currently, there is no treatment for TBI-induced cell damage and neural circuit dysfunction in the brain, and thus, it is important to understand the underlying cellular mechanisms that lead to cell damage. In the current study, we use stretchable microelectrode arrays (sMEAs) to model the primary injury of TBI to study the electrophysiological effects of physically injuring cortical cells. We recorded electrophysiological activity before injury and then stretched the flexible membrane of the sMEAs to injure the cells to varying degrees. At 1, 24, and 72 h post-stretch, we recorded activity to analyze differences in spike rate, Fano factor, burstlet rate, burstlet width, synchrony of firing, local network efficiency, and Q statistic. Our results demonstrate that mechanical injury changes the firing properties of cortical neuron networks in culture in a time- and severity-dependent manner. Our results suggest that changes to electrophysiological properties after stretch are dependent on the strength of synchronization between neurons prior to injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Humanos , Neuronas/fisiología , Encéfalo
17.
Glia ; 61(3): 394-408, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23281078

RESUMEN

Congenital hyperinsulinism/hyperammonemia (HI/HA) syndrome is caused by an activation mutation of glutamate dehydrogenase 1 (GDH1), a mitochondrial enzyme responsible for the reversible interconversion between glutamate and α-ketoglutarate. The syndrome presents clinically with hyperammonemia, significant episodic hypoglycemia, seizures, and frequent incidences of developmental and learning defects. Clinical research has implicated that although some of the developmental and neurological defects may be attributed to hypoglycemia, some characteristics cannot be ascribed to low glucose and as hyperammonemia is generally mild and asymptomatic, there exists the possibility that altered GDH1 activity within the brain leads to some clinical changes. GDH1 is allosterically regulated by many factors, and has been shown to be inhibited by the ADP-ribosyltransferase sirtuin 4 (SIRT4), a mitochondrially localized sirtuin. Here we show that SIRT4 is localized to mitochondria within the brain. SIRT4 is highly expressed in glial cells, specifically astrocytes, in the postnatal brain and in radial glia during embryogenesis. Furthermore, SIRT4 protein decreases in expression during development. We show that factors known to allosterically regulate GDH1 alter gliogenesis in CTX8 cells, a novel radial glial cell line. We find that SIRT4 and GDH1 overexpression play antagonistic roles in regulating gliogenesis and that a mutant variant of GDH1 found in HI/HA patients accelerates the development of glia from cultured radial glia cells.


Asunto(s)
Corteza Cerebral/metabolismo , Glutamato Deshidrogenasa/metabolismo , Neuroglía/metabolismo , Sirtuinas/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Glutamato Deshidrogenasa/genética , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hipoglucemia/genética , Hipoglucemia/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Neuroglía/citología , Ratas , Sirtuinas/genética
18.
Mol Cell Neurosci ; 50(1): 10-20, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22465229

RESUMEN

Dendrite morphogenesis is a complex but well-orchestrated process and includes the development of dendritic branches, forming characteristic dendrite arbors, and dendritic spines, allowing neurons to communicate with each other. Various studies report that many neuropsychiatric disorders are characterized by dendritic and synaptic pathology, including abnormal spine density and morphology, synapse loss, and aberrant synaptic signaling and plasticity. In this review, we discuss dendrite development and branching, and in specific, morphology, cytoskeletal architecture, and how the complexity of the dendrite tree and its functional capabilities are altered in various brain disorders. Identifying and understanding these changes in dendrite morphology are essential for understanding brain function in normal and disease states.


Asunto(s)
Encéfalo/patología , Dendritas/patología , Trastornos Mentales/patología , Animales , Axones/metabolismo , Axones/patología , Encéfalo/crecimiento & desarrollo , Citoesqueleto/metabolismo , Dendritas/metabolismo , Humanos , Ratones , Enfermedades del Sistema Nervioso/patología , Sinapsis/metabolismo , Sinapsis/patología
19.
Pharmacol Res Perspect ; 11(3): e01071, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37133236

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting Coronavirus disease 2019 emerged in late 2019 and is responsible for significant morbidity and mortality worldwide. A hallmark of severe COVID-19 is exaggerated systemic inflammation, regarded as a "cytokine storm," which contributes to the damage of various organs, primarily the lungs. The inflammation associated with some viral illnesses is known to alter the expression of drug-metabolizing enzymes and transporters. These alterations can lead to modifications in drug exposure and the processing of various endogenous compounds. Here, we provide evidence to support changes in the mitochondrial ribonucleic acid expression of a subset of drug transporters (84 transporters) in the liver, kidneys, and lungs and metabolizing enzymes (84 enzymes) in the liver in a humanized angiotensin-converting enzyme 2 receptor mouse model. Specifically, three drug transporters (Abca3, Slc7a8, Tap1) and the pro-inflammatory cytokine IL-6 were upregulated in the lungs of SARS-CoV-2 infected mice. We also found significant downregulation of drug transporters responsible for the movement of xenobiotics in the liver and kidney. Additionally, expression of cytochrome P-450 2f2 which is known to metabolize some pulmonary toxicants, was significantly decreased in the liver of infected mice. The significance of these findings requires further exploration. Our results suggest that further research should emphasize altered drug disposition when investigating therapeutic compounds, whether re-purposed or new chemical entities, in other animal models and ultimately in individuals infected with SARS-CoV-2. Moreover, the influence and impact of these changes on the processing of endogenous compounds also require further investigation.


Asunto(s)
COVID-19 , Ratones , Animales , SARS-CoV-2 , Modelos Animales de Enfermedad , Peptidil-Dipeptidasa A/metabolismo , Inflamación
20.
Commun Biol ; 6(1): 1278, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110605

RESUMEN

Plasticity and homeostatic mechanisms allow neural networks to maintain proper function while responding to physiological challenges. Despite previous work investigating morphological and synaptic effects of brain-derived neurotrophic factor (BDNF), the most prevalent growth factor in the central nervous system, how exposure to BDNF manifests at the network level remains unknown. Here we report that BDNF treatment affects rodent hippocampal network dynamics during development and recovery from glutamate-induced excitotoxicity in culture. Importantly, these effects are not obvious when traditional activity metrics are used, so we delve more deeply into network organization, functional analyses, and in silico simulations. We demonstrate that BDNF partially restores homeostasis by promoting recovery of weak and medium connections after injury. Imaging and computational analyses suggest these effects are caused by changes to inhibitory neurons and connections. From our in silico simulations, we find that BDNF remodels the network by indirectly strengthening weak excitatory synapses after injury. Ultimately, our findings may explain the difficulties encountered in preclinical and clinical trials with BDNF and also offer information for future trials to consider.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Sinapsis , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Sinapsis/metabolismo , Neuronas/fisiología , Ácido Glutámico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA