Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurochem ; 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38994820

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive phospholipid that participates in critical processes in neural development and adult brain function and is implicated in various pathophysiological conditions. Along with its six well-characterized receptors, atypical regulators of LPA signaling have also been suggested, including phospholipid phosphatase-related proteins (PLPPRs). PLPPRs have been mostly studied in the developing brain where they control LPA-dependent axon guidance, cortical network hyperexcitability, and glutamatergic neurotransmission. PLPPR4 and PLPPR3 represent two closely related proteins reported to localize predominantly in dendrites and axons, respectively, and differ in their developmental expression patterns. Herein, we have revised the expression patterns of PLPPRs in the cerebellum, dorsal and ventral hippocampus, prefrontal cortex (PFC), nucleus accumbens, and striatum during development and in the adult using quantitative PCR. Expression patterns of Plppr2,4 and 5 were consistent with previous studies, whereas Plppr3 and Plppr1 exhibited a unique expression profile in nucleus accumbens (NAc) and striatum in later developmental and adult stages, which we verified at the protein level for PLPPR3. To investigate neuron type-specific expression at the single cell level, we developed a bioinformatic tool to analyze recent single-cell RNA-sequencing data in the cerebral cortex and hippocampus of adult mice. Our analysis revealed a widespread but also selective adult neuron-type expression with higher expression levels of Plppr3, Plppr1, and Plppr5 in GABAergic and Plppr4 and Plppr2 in glutamatergic neurons. PLPPR4 has been identified as a post-synaptic modulator of LPA levels in glutamatergic synapses operating via an uptake mechanism, to control LPA-dependent cortical network hyperexcitability. Using subcellular fractionation experiments, we found that both PLPPR4 and PLPPR3 are co-expressed in adult synaptosomal membranes. Furthermore, flow cytometry experiments in HEK293 cells showed comparable LPA uptake by PLPPR4 and PLPPR3, whereas PLPRR3, but not PLPPR4, induced also uptake of monoacylglycerol, the dephosphorylation product of LPA. We propose that synaptic LPA may be subject to both pre-synaptic and post-synaptic mechanisms of regulation by PLPPRs in addition to LPARs in developing and adult synapses.

2.
Nanoscale Adv ; 6(15): 3825-3837, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39050941

RESUMEN

Citrate-coated iron oxide nanoparticles, specifically Synomag®-COOH (SynC), are promising tracers in magnetic particle imaging (MPI) due to their high magnetic moments and rapid cellular uptake. The mechanisms driving efficient SynC uptake remain unclear. Previous observations suggest a role of the extracellular glycocalyx during nanoparticle uptake. Here, we ascertain whether the cell-surface glycosaminoglycans (GAGs) regulate the uptake of SynC. Using transmission electron microscopy (TEM), we visualized SynC uptake by THP-1 cells, a human acute monocytic leukemia cell line. We investigated the interaction of SynC with GAGs in living cells using click-chemistry-based labeling. Upon treating THP-1 cells with chondroitinase or hyaluronidase and with a xylosyltransferase-deficient cell line, we quantified SynC uptake and measured interactions of SynC with cells in real time using magnetic particle spectroscopy (MPS). The THP-1 cell membrane engulfed or formed extensions around SynC, indicating uptake through pinocytosis and phagocytosis. We measured an increased MPS signal of SynC within seconds of cell contact, suggesting an interaction with extracellular components like the glycocalyx. Upon adding SynC to THP-1 cells, we could not observe disruption of fluorescently labeled GAGs or an enhanced intracellular fluorescence, implying that SynC does not accelerate the turnover of GAGs by binding. Lack of chondroitin sulfate, heparan sulfate, and hyaluronic acid did not affect the rapid magnetic behavior increase of SynC upon cell contact. Accordingly, we measured no significant differences in SynC uptake between wild type cells and our GAG-deficient models. These findings suggest that GAGs act as a permeable bandpass for SynC nanoparticles with a minor negative surface charge of -13.8 mV. This finding has significant implications for MPI-based cell tracking because it facilitates efficient tracking of cell types that lack a strong repulsion by cell-surface GAGs. It will be crucial to investigate whether the rapid uptake of SynC is cell-type specific and influenced by different extracellular matrix compositions.

3.
Nat Commun ; 15(1): 4879, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849354

RESUMEN

The mammalian neocortex comprises an enormous diversity regarding cell types, morphology, and connectivity. In this work, we discover a post-transcriptional mechanism of gene expression regulation, protein translation, as a determinant of cortical neuron identity. We find specific upregulation of protein synthesis in the progenitors of later-born neurons and show that translation rates and concomitantly protein half-lives are inherent features of cortical neuron subtypes. In a small molecule screening, we identify Ire1α as a regulator of Satb2 expression and neuronal polarity. In the developing brain, Ire1α regulates global translation rates, coordinates ribosome traffic, and the expression of eIF4A1. Furthermore, we demonstrate that the Satb2 mRNA translation requires eIF4A1 helicase activity towards its 5'-untranslated region. Altogether, we show that cortical neuron diversity is generated by mechanisms operating beyond gene transcription, with Ire1α-safeguarded proteostasis serving as an essential regulator of brain development.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz , Neocórtex , Neuronas , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas , Animales , Neocórtex/metabolismo , Neocórtex/citología , Neocórtex/embriología , Neuronas/metabolismo , Neuronas/citología , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Regulación del Desarrollo de la Expresión Génica , Proteostasis , Neurogénesis/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Regiones no Traducidas 5'/genética , Ribosomas/metabolismo , Ribosomas/genética , Humanos , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Diferenciación Celular/genética
4.
Brain Behav Immun ; 117: 399-411, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38309639

RESUMEN

BACKGROUND: Autoantibodies against the potassium voltage-gated channel subfamily A member 2 (KCNA2) have been described in a few cases of neuropsychiatric disorders, but their diagnostic and pathophysiological role is currently unknown, imposing challenges to medical practice. DESIGN / METHODS: We retrospectively collected comprehensive clinical and paraclinical data of 35 patients with KCNA2 IgG autoantibodies detected in cell-based and tissue-based assays. Patients' sera and cerebrospinal fluid (CSF) were used for characterization of the antigen, clinical-serological correlations, and determination of IgG subclasses. RESULTS: KCNA2 autoantibody-positive patients (n = 35, median age at disease onset of 65 years, range of 16-83 years, 74 % male) mostly presented with cognitive impairment and/or epileptic seizures but also ataxia, gait disorder and personality changes. Serum autoantibodies belonged to IgG3 and IgG1 subclasses and titers ranged from 1:32 to 1:10,000. KCNA2 IgG was found in the CSF of 8/21 (38 %) patients and in the serum of 4/96 (4.2 %) healthy blood donors. KCNA2 autoantibodies bound to characteristic anatomical areas in the cerebellum and hippocampus of mammalian brain and juxtaparanodal regions of peripheral nerves but reacted exclusively with intracellular epitopes. A subset of four KCNA2 autoantibody-positive patients responded markedly to immunotherapy alongside with conversion to seronegativity, in particular those presenting an autoimmune encephalitis phenotype and receiving early immunotherapy. An available brain biopsy showed strong immune cell invasion. KCNA2 autoantibodies occurred in less than 10 % in association with an underlying tumor. CONCLUSION: Our data suggest that KCNA2 autoimmunity is clinically heterogeneous. Future studies should determine whether KCNA2 autoantibodies are directly pathogenic or develop secondarily. Early immunotherapy should be considered, in particular if autoantibodies occur in CSF or if clinical or diagnostic findings suggest ongoing inflammation. Suspicious clinical phenotypes include autoimmune encephalitis, atypical dementia, new-onset epilepsy and unexplained epileptic seizures.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Autoinmunidad , Encefalitis , Enfermedad de Hashimoto , Animales , Humanos , Masculino , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Femenino , Estudios Retrospectivos , Autoanticuerpos , Convulsiones , Mamíferos , Canal de Potasio Kv.1.2
5.
Brain Behav Immun Health ; 33: 100678, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37692096

RESUMEN

Anti-neuronal autoantibodies can be transplacentally transferred during pregnancy and may cause detrimental effects on fetal development. It is unclear whether autoantibodies against synapsin-I, one of the most abundant synaptic proteins, are associated with developmental abnormalities in humans. We recruited a cohort of 263 pregnant women and detected serum synapsin-I IgG autoantibodies in 13.3% using cell-based assays. Seropositivity was strongly associated with abnormalities of fetal development including structural defects, intrauterine growth retardation, amniotic fluid disorders and neuropsychiatric developmental diseases in previous children (odds ratios of 3-6.5). Autoantibodies reached the fetal circulation and were mainly of IgG1/IgG3 subclasses. They bound to conformational and linear synapsin-I epitopes, five distinct epitopes were identified using peptide microarrays. The findings indicate that synapsin-I autoantibodies may be clinically useful biomarkers or even directly participate in the disease process of neurodevelopmental disorders, thus being potentially amenable to antibody-targeting interventional strategies in the future.

6.
J Exp Med ; 218(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34546336

RESUMEN

Autoantibodies targeting the GABAA receptor (GABAAR) hallmark an autoimmune encephalitis presenting with frequent seizures and psychomotor abnormalities. Their pathogenic role is still not well-defined, given the common overlap with further autoantibodies and the lack of patient-derived mAbs. Five GABAAR mAbs from cerebrospinal fluid cells bound to various epitopes involving the α1 and γ2 receptor subunits, with variable binding strength and partial competition. mAbs selectively reduced GABAergic currents in neuronal cultures without causing receptor internalization. Cerebroventricular infusion of GABAAR mAbs and Fab fragments into rodents induced a severe phenotype with seizures and increased mortality, reminiscent of encephalitis patients' symptoms. Our results demonstrate direct pathogenicity of autoantibodies on GABAARs independent of Fc-mediated effector functions and provide an animal model for GABAAR encephalitis. They further provide the scientific rationale for clinical treatments using antibody depletion and can serve as tools for the development of antibody-selective immunotherapies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Autoanticuerpos/inmunología , Encefalitis/inmunología , Epilepsia/inmunología , Receptores de GABA-A/inmunología , Convulsiones/inmunología , Animales , Autoantígenos/inmunología , Células Cultivadas , Células HEK293 , Hipocampo/inmunología , Humanos , Ratones , Neuronas/inmunología
7.
Mol Cell ; 81(2): 304-322.e16, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33357414

RESUMEN

Protein synthesis must be finely tuned in the developing nervous system as the final essential step of gene expression. This study investigates the architecture of ribosomes from the neocortex during neurogenesis, revealing Ebp1 as a high-occupancy 60S peptide tunnel exit (TE) factor during protein synthesis at near-atomic resolution by cryoelectron microscopy (cryo-EM). Ribosome profiling demonstrated Ebp1-60S binding is highest during start codon initiation and N-terminal peptide elongation, regulating ribosome occupancy of these codons. Membrane-targeting domains emerging from the 60S tunnel, which recruit SRP/Sec61 to the shared binding site, displace Ebp1. Ebp1 is particularly abundant in the early-born neural stem cell (NSC) lineage and regulates neuronal morphology. Ebp1 especially impacts the synthesis of membrane-targeted cell adhesion molecules (CAMs), measured by pulsed stable isotope labeling by amino acids in cell culture (pSILAC)/bioorthogonal noncanonical amino acid tagging (BONCAT) mass spectrometry (MS). Therefore, Ebp1 is a central component of protein synthesis, and the ribosome TE is a focal point of gene expression control in the molecular specification of neuronal morphology during development.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Neocórtex/metabolismo , Neuronas/metabolismo , Biosíntesis de Proteínas , Proteostasis/genética , Proteínas de Unión al ARN/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética , Animales , Animales Recién Nacidos , Sitios de Unión , Moléculas de Adhesión Celular Neuronal/química , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular Tumoral , Microscopía por Crioelectrón , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Masculino , Ratones , Neocórtex/citología , Neocórtex/crecimiento & desarrollo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/citología , Cultivo Primario de Células , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/ultraestructura , Partícula de Reconocimiento de Señal/química , Partícula de Reconocimiento de Señal/genética , Partícula de Reconocimiento de Señal/metabolismo
8.
Front Cell Neurosci ; 14: 596072, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240046

RESUMEN

The current study investigates the neurotrophic effects of Clostridium botulinum C3 transferase (C3bot) on highly purified, glia-free, GABAergic, and glutamatergic neurons. Incubation with nanomolar concentrations of C3bot promotes dendrite formation as well as dendritic and axonal outgrowth in rat GABAergic neurons. A comparison of C3bot effects on sorted mouse GABAergic and glutamatergic neurons obtained from newly established NexCre;Ai9xVGAT Venus mice revealed a higher sensitivity of GABAergic cells to axonotrophic and dendritic effects of C3bot in terms of process length and branch formation. Protein biochemical analysis of known C3bot binding partners revealed comparable amounts of ß1 integrin in both cell types but a higher expression of vimentin in GABAergic neurons. Accordingly, binding of C3bot to GABAergic neurons was stronger than binding to glutamatergic neurons. A combinatory treatment of glutamatergic neurons with C3bot and vimentin raised the amount of bound C3bot to levels comparable to the ones in GABAergic neurons, thereby confirming the specificity of effects. Overall, different surface vimentin levels between GABAergic and glutamatergic neurons exist that mediate neurotrophic C3bot effects.

9.
Int J Mol Sci ; 21(5)2020 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-32106593

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a major neuronal growth factor that is widely expressed in the central nervous system. It is synthesized as a glycosylated precursor protein, (pro)BDNF and post-translationally converted to the mature form, (m)BDNF. BDNF is known to be produced and secreted by cortical glutamatergic principal cells (PCs); however, it remains a question whether it can also be synthesized by other neuron types, in particular, GABAergic interneurons (INs). Therefore, we utilized immunocytochemical labeling and reverse transcription quantitative PCR (RT-qPCR) to investigate the cellular distribution of proBDNF and its RNA in glutamatergic and GABAergic neurons of the mouse cortex. Immunofluorescence labeling revealed that mBDNF, as well as proBDNF, localized to both the neuronal populations in the hippocampus. The precursor proBDNF protein showed a perinuclear distribution pattern, overlapping with the rough endoplasmic reticulum (ER), the site of protein synthesis. RT-qPCR of samples obtained using laser capture microdissection (LCM) or fluorescence-activated cell sorting (FACS) of hippocampal and cortical neurons further demonstrated the abundance of BDNF transcripts in both glutamatergic and GABAergic cells. Thus, our data provide compelling evidence that BDNF can be synthesized by both principal cells and INs of the cortex.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Corteza Cerebral/metabolismo , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Masculino , Ratones
10.
Artículo en Inglés | MEDLINE | ID: mdl-31178713

RESUMEN

The advent of optogenetic methods has made it possible to use endogeneously produced molecules to image and manipulate cellular, subcellular, and synaptic activity. It has also led to the development of photoactivatable calcium-dependent indicators that mark active synapses, neurons, and circuits. Furthermore, calcium-dependent photoactivation can be used to trigger gene expression in active neurons. Here we describe two sets of protocols, one using CaMPARI and a second one using Cal-Light. CaMPARI, a calcium-modulated photoactivatable ratiometric integrator, enables rapid network-wide, tunable, all-optical functional circuit mapping. Cal-Light, a photoactivatable calcium sensor, while slower to respond than CaMPARI, has the capacity to trigger the expression of genes, including effectors, activators, indicators, or other constructs. Here we describe the rationale and provide procedures for using these two calcium-dependent constructs (1) in vitro in dissociated primary neuronal cell cultures (CaMPARI & Cal-Light); (2) in vitro in acute brain slices for circuit mapping (CaMPARI); (3) in vivo for triggering photoconversion or gene expression (CaMPARI & Cal-Light); and finally, (4) for recovering photoconverted neurons post-fixation with immunocytochemistry (CaMPARI). The approaches and protocols we describe are examples of the potential uses of both CaMPARI & Cal-Light. The ability to mark and manipulate neurons that are active during specific epochs of behavior has a vast unexplored experimental potential.

11.
J Vis Exp ; (148)2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31233034

RESUMEN

The overall goal of this protocol is to generate purified neuronal cultures derived from either GABAergic or glutamatergic neurons. Purified neurons can be cultured in defined media for 16 days in vitro and are amenable to any analyses typically performed on dissociated cultures, including electrophysiological, morphological and survival analyses. The major advantage of these cultures is that specific cell types can be selectively studied in the absence of complex external influences, such as those arising from glial cells or other neuron types. When planning experiments with purified cells, however, it is important to note that neurons strongly depend on glia-conditioned media for their growth and survival. In addition, glutamatergic neurons further depend on glia-secreted factors for the establishment of synaptic transmission. We therefore also describe a method for co-culturing neurons and glial cells in a non-contact arrangement. Using these methods, we have identified major differences between the development of GABAergic and glutamatergic neuronal networks. Thus, studying cultures of purified neurons has great potential for furthering our understanding of how the nervous system develops and functions. Moreover, purified cultures may be useful for investigating the direct action of pharmacological agents, growth factors or for exploring the consequences of genetic manipulations on specific cell types. As more and more transgenic animals become available, labeling additional specific cell types of interest, we expect that the protocols described here will grow in their applicability and potential.


Asunto(s)
Citometría de Flujo , Neuronas/fisiología , Cultivo Primario de Células/métodos , Animales , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Ácido Glutámico/metabolismo , Neuroglía/citología , Neuronas/clasificación , Transmisión Sináptica , Ácido gamma-Aminobutírico/metabolismo
12.
Cereb Cortex ; 29(3): 1230-1243, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29425353

RESUMEN

In the mammalian cortex, GABAergic and glutamatergic neurons represent 2 major neuronal classes, which establish inhibitory and excitatory synapses, respectively. Despite differences in their anatomy, physiology and developmental origin, both cell types require support from glial cells, particularly astrocytes, for their growth and survival. Recent experiments indicate that glutamatergic neurons also depend on astrocytes for synapse formation. However, it is not clear if the same holds true for GABAergic neurons. By studying highly pure GABAergic cell cultures, established through fluorescent activated cell sorting, we find that purified GABAergic neurons are smaller and have reduced survival, nevertheless they establish robust synaptic transmission in the absence of glia. Support from glial cells reverses morphological and survival deficits, but does little to alter synaptic transmission. In contrast, in cultures of purified glutamatergic neurons, morphological development, survival and synaptic transmission are collectively dependent on glial support. Thus, our results demonstrate a fundamental difference in the way GABAergic and glutamatergic neurons depend on glia for the establishment of synaptic transmission, a finding that has important implications for our understanding of how neuronal networks develop.


Asunto(s)
Neuronas GABAérgicas/fisiología , Ácido Glutámico/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Transmisión Sináptica , Potenciales de Acción , Animales , Células Cultivadas , Femenino , Neuronas GABAérgicas/citología , Ratones Endogámicos C57BL , Neuroglía/citología , Neuronas/citología , Ratas Wistar , Sinapsis/fisiología
13.
Brain Res ; 1704: 249-256, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30347219

RESUMEN

The y-aminobutyric acid A receptor (GABAAR) participates in most neurophysiological processes. Mutations cause epilepsy and neuropsychiatric pathologies. Recently a severe encephalitis with refractory seizures and antibodies against GABAARs has been described. Considering the complex subunit distribution of GABAARs, binding patterns of human GABAAR antibodies will help to understand the pathophysiology underlying diverse clinical pictures. We therefore investigated the cerebrospinal fluid (CSF) reactivity of a patient with GABAAR encephalitis using immunocytochemistry on murine brain sections and compared its specificity with commercial GABAAR antibodies. The immunoreactivity of the patient's CSF showed excellent agreement with previously reported GABAAR mRNA expression. Colocalization with neuronal and glial markers verified the neuronal specificity of GABAAR. Patient antibodies strongly bound to neuropil in the external layers of the olfactory bulb, CA1 and CA2 of the hippocampus, neocortex, pallidum and granular cells of the cerebellum. Distribution patterns suggest the presence of polyclonal CSF GABAAR antibodies targeting multiple receptor subunits. The comparison with commercial antibodies revealed large overlap, but also specific differences. For example, commercial antibodies accumulated on dendrites, while CSF created a homogeneous neuropil signal. The number of GABAergic synapses stained with CSF exceeded those labeled with the commercial antibodies. In some areas, commercial antibodies and CSF even stained complementary populations of GABAergic neurons. The data indicate the presence of additional anti-neuronal autoantibodies in the CSF, which could be assessed in future studies with individual recombinant monoclonal antibodies from CSF B cells. This strategy would confirm antibody pathogenicity and likely explain variable clinical pictures in autoimmune encephalitis patients.


Asunto(s)
Autoanticuerpos/metabolismo , Corteza Cerebral/metabolismo , Encefalitis/inmunología , Hipocampo/metabolismo , Receptores de GABA-A/inmunología , Animales , Niño , Encefalitis/metabolismo , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratas , Ratas Wistar , Receptores de GABA-A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA