Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(10): 113175, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37756163

RESUMEN

The mechanical properties of solid tumors influence tumor cell phenotype and the ability to invade surrounding tissues. Using bioengineered scaffolds to provide a matrix microenvironment for patient-derived glioblastoma (GBM) spheroids, this study demonstrates that a soft, brain-like matrix induces GBM cells to shift to a glycolysis-weighted metabolic state, which supports invasive behavior. We first show that orthotopic murine GBM tumors are stiffer than peritumoral brain tissues, but tumor stiffness is heterogeneous where tumor edges are softer than the tumor core. We then developed 3D scaffolds with µ-compressive moduli resembling either stiffer tumor core or softer peritumoral brain tissue. We demonstrate that the softer matrix microenvironment induces a shift in GBM cell metabolism toward glycolysis, which manifests in lower proliferation rate and increased migration activities. Finally, we show that these mechanical cues are transduced from the matrix via CD44 and integrin receptors to induce metabolic and phenotypic changes in cancer cells.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Animales , Ratones , Glioblastoma/patología , Línea Celular Tumoral , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Microambiente Tumoral
2.
Exp Neurol ; 362: 114339, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36717013

RESUMEN

Large scale studies in populations of European and Han Chinese ancestry found a series of rare gain-of-function microduplications in VIPR2, encoding VPAC2, a receptor that binds vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide with high affinity, that were associated with an up to 13-fold increased risk for schizophrenia. To address how VPAC2 receptor overactivity might affect brain development, we used a well-characterized Nestin-Cre mouse strain and a knock-in approach to overexpress human VPAC2 in the central nervous system. Mice that overexpressed VPAC2 were found to exhibit a significant reduction in brain weight. Magnetic resonance imaging analysis confirmed a decrease in brain size, a specific reduction in the hippocampus grey matter volume and a paradoxical increase in whole-brain white matter volume. Sex-specific changes in behavior such as impaired prepulse inhibition and contextual fear memory were observed in VPAC2 overexpressing mice. The data indicate that the VPAC2 receptor may play a critical role in brain morphogenesis and suggest that overactive VPAC2 signaling during development plays a mechanistic role in some forms of schizophrenia.


Asunto(s)
Receptores de Tipo II del Péptido Intestinal Vasoactivo , Sustancia Blanca , Masculino , Humanos , Femenino , Ratones , Animales , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Sustancia Blanca/metabolismo , Péptido Intestinal Vasoactivo/química , Péptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Inhibición Prepulso
3.
Adv Healthc Mater ; 12(14): e2203143, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36694362

RESUMEN

Increased secretion of hyaluronic acid (HA), a glycosaminoglycan abundant in the brain extracellular matrix (ECM), correlates with worse clinical outcomes for glioblastoma (GBM) patients. GBM cells aggressively invade the brain parenchyma while encountering spatiotemporal changes in their local ECM, including HA concentration. To investigate how varying HA concentrations affect GBM invasion, patient-derived GBM cells are cultured within a soft, 3D matrix in which HA concentration is precisely varied and cell migration observed. Data demonstrate that HA concentration can determine the invasive activity of patient-derived GBM cells in a biphasic and highly sensitive manner, where the absolute concentration of HA at which cell migration peaked is specific to each patient-derived line. Furthermore, evidence that this response relies on phosphorylated ezrin, which interacts with the intracellular domain of HA-engaged CD44 to effectively link the actin cytoskeleton to the local ECM is provided. Overall, this study highlights CD44-HA binding as a major mediator of GBM cell migration that acts independently of integrins and focal adhesion complexes and suggests that targeting HA-CD44-ezrin interactions represents a promising therapeutic strategy to prevent tumor cell invasion in the brain.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/patología , Ácido Hialurónico/química , Línea Celular Tumoral , Encéfalo/patología , Movimiento Celular , Receptores de Hialuranos/metabolismo
4.
Cell Rep ; 41(3): 111511, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36261010

RESUMEN

Glioblastoma (GBM) is characterized by extensive microvascular hyperproliferation. In addition to supplying blood to the tumor, GBM vessels also provide trophic support to glioma cells and serve as conduits for migration into the surrounding brain, promoting recurrence. Here, we enrich CD31-expressing glioma vascular cells (GVCs) and A2B5-expressing glioma tumor cells (GTCs) from primary GBM and use RNA sequencing to create a comprehensive molecular interaction map of the secreted and extracellular factors elaborated by GVCs that can interact with receptors and membrane molecules on GTCs. To validate our findings, we utilize functional assays, including a hydrogel-based migration assay and in vivo mouse models to demonstrate that one identified factor, the little-studied integrin binding sialoprotein (IBSP), enhances tumor growth and promotes the migration of GTCs along the vasculature. This perivascular niche interactome will serve as a resource to the research community in defining the potential functions of the GBM vasculature.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Ratones , Glioblastoma/patología , Sialoproteína de Unión a Integrina/metabolismo , Neoplasias Encefálicas/patología , Células Madre Neoplásicas/metabolismo , Glioma/patología , Movimiento Celular , Hidrogeles
5.
Nat Commun ; 13(1): 6202, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36261421

RESUMEN

Glioma stem cells (GSC) exhibit plasticity in response to environmental and therapeutic stress leading to tumor recurrence, but the underlying mechanisms remain largely unknown. Here, we employ single-cell and whole transcriptomic analyses to uncover that radiation induces a dynamic shift in functional states of glioma cells allowing for acquisition of vascular endothelial-like and pericyte-like cell phenotypes. These vascular-like cells provide trophic support to promote proliferation of tumor cells, and their selective depletion results in reduced tumor growth post-treatment in vivo. Mechanistically, the acquisition of vascular-like phenotype is driven by increased chromatin accessibility and H3K27 acetylation in specific vascular genes allowing for their increased expression post-treatment. Blocking P300 histone acetyltransferase activity reverses the epigenetic changes induced by radiation and inhibits the adaptive conversion of GSC into vascular-like cells and tumor growth. Our findings highlight a role for P300 in radiation-induced stress response, suggesting a therapeutic approach to prevent glioma recurrence.


Asunto(s)
Glioma , Recurrencia Local de Neoplasia , Humanos , Recurrencia Local de Neoplasia/patología , Glioma/genética , Glioma/radioterapia , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Cromatina/metabolismo , Histona Acetiltransferasas/metabolismo
6.
Neurobiol Dis ; 160: 105524, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34610465

RESUMEN

Chronic inflammation drives synaptic loss in multiple sclerosis (MS) and is also commonly observed in other neurodegenerative diseases. Clinically approved treatments for MS provide symptomatic relief but fail to halt neurodegeneration and neurological decline. Studies in animal disease models have demonstrated that the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP, ADCYAP1) exhibits anti-inflammatory, neuroprotective and regenerative properties. Anti-inflammatory actions appear to be mediated primarily by two receptors, VPAC1 and VPAC2, which also bind vasoactive intestinal peptide (VIP). Pharmacological experiments indicate that another receptor, PAC1 (ADCYAP1R1), which is highly selective for PACAP, provides protection to neurons, although genetic evidence and other mechanistic information is lacking. To determine if PAC1 receptors protect neurons in a cell-autonomous manner, we used adeno-associated virus (AAV2) to deliver Cre recombinase to the retina of mice harboring floxed PAC1 alleles. Mice were then subjected to chronic experimental autoimmune encephalomyelitis (EAE), a disease model that recapitulates major clinical and pathological features of MS and associated optic neuritis. Unexpectedly, deletion of PAC1 in naïve mice resulted in a deficit of retinal ganglionic neurons (RGNs) and their dendrites, suggesting a homeostatic role of PAC1. Moreover, deletion of PAC1 resulted in increased EAE-induced loss of a subpopulation of RGNs purported to be vulnerable in animal models of glaucoma. Increased axonal pathology and increased secondary presence of microglia/macrophages was also prominently seen in the optic nerve. These findings demonstrate that neuronal PAC1 receptors play a homeostatic role in protecting RGNs and directly protects neurons and their axons against neuroinflammatory challenge. SIGNIFICANCE STATEMENT: Chronic inflammation is a major component of neurodegenerative diseases and plays a central role in multiple sclerosis (MS). Current treatments for MS do not prevent neurodegeneration and/or neurological decline. The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to have anti-inflammatory, neuroprotective and regenerative properties but the cell type- and receptor-specific mechanisms are not clear. To test whether the protective effects of PACAP are direct on the PAC1 receptor subtype on neurons, we delete PAC1 receptors from neurons and investigate neuropathologigical changes in an animal model of MS. The findings demonstrate that PAC1 receptors on neurons play a homeostatic role in maintaining neuron health and can directly protect neurons and their axons during neuroinflammatory disease.


Asunto(s)
Axones/metabolismo , Muerte Celular/fisiología , Esclerosis Múltiple/metabolismo , Neuritis Óptica/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Neuronas Retinianas/metabolismo , Animales , Axones/patología , Encéfalo/metabolismo , Encéfalo/patología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Neuritis Óptica/genética , Neuritis Óptica/patología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética
7.
Nat Commun ; 12(1): 3958, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172753

RESUMEN

Astrocytes play important roles in neurological disorders such as stroke, injury, and neurodegeneration. Most knowledge on astrocyte biology is based on studies of mouse models and the similarities and differences between human and mouse astrocytes are insufficiently characterized, presenting a barrier in translational research. Based on analyses of acutely purified astrocytes, serum-free cultures of primary astrocytes, and xenografted chimeric mice, we find extensive conservation in astrocytic gene expression between human and mouse samples. However, the genes involved in defense response and metabolism show species-specific differences. Human astrocytes exhibit greater susceptibility to oxidative stress than mouse astrocytes, due to differences in mitochondrial physiology and detoxification pathways. In addition, we find that mouse but not human astrocytes activate a molecular program for neural repair under hypoxia, whereas human but not mouse astrocytes activate the antigen presentation pathway under inflammatory conditions. Here, we show species-dependent properties of astrocytes, which can be informative for improving translation from mouse models to humans.


Asunto(s)
Astrocitos/fisiología , Animales , Presentación de Antígeno , Astrocitos/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Inactivación Metabólica , Inflamación , Ratones , Mitocondrias/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/patología , Estrés Oxidativo , Poli I-C/farmacología , Poli I-C/uso terapéutico , Especificidad de la Especie , Transcriptoma/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/uso terapéutico
8.
iScience ; 23(9): 101453, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32861192

RESUMEN

Glioblastoma (GBM) metabolism has traditionally been characterized by a primary dependence on aerobic glycolysis, prompting the use of the ketogenic diet (KD) as a potential therapy. In this study we evaluated the effectiveness of the KD in GBM and assessed the role of fatty acid oxidation (FAO) in promoting GBM propagation. In vitro assays revealed FA utilization throughout the GBM metabolome and growth inhibition in nearly every cell line in a broad spectrum of patient-derived glioma cells treated with FAO inhibitors. In vivo assessments revealed that knockdown of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme for FAO, reduced the rate of tumor growth and increased survival. However, the unrestricted ketogenic diet did not reduce tumor growth and for some models significantly reduced survival. Altogether, these data highlight important roles for FA and ketone body metabolism that could serve to improve targeted therapies in GBM.

9.
J Mol Neurosci ; 68(3): 439-451, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30058008

RESUMEN

The sympathetic nervous system (SNS) serves to maintain homeostasis of vital organ systems throughout the body, and its dysfunction plays a major role in human disease. The SNS also links the central nervous system to the immune system during different types of stress via innervation of the lymph nodes, spleen, thymus, and bone marrow. Previous studies have shown that pituitary adenylate cyclase-activating polypeptide (PACAP, gene name adcyap1) exhibits anti-inflammatory properties in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Because PACAP is known to regulate SNS function, we hypothesized that part of the immunoprotective action of PACAP is due to its neuromodulatory effects on sympathetic neurons. To examine this, we used an inducible, targeted approach to conditionally disrupt not only the PACAP-preferring PAC1 receptor gene (adcyap1r1) in dopamine ß-hydroxylase-expressing cells, which includes postganglionic sympathetic neurons, but also catecholaminergic neurons in the brain and adrenomedullary chromaffin cells. In contrast to our previous EAE studies using PACAP global knockout mice which developed severe and prolonged EAE, we found that mice with conditional loss of PAC1 receptors in catecholaminergic cells developed a delayed time course of EAE with reduced helper T cell type 1 (Th1) and Th17 and enhanced Th2 cell polarization. At later time points, similar to mice with global PACAP loss, mice with conditional loss of PAC1 exhibited more severe clinical disease than controls. The latter was associated with a reduction in the abundance of thymic regulatory T cells (Tregs). These studies indicate that PAC1 receptor signaling acts in catecholaminergic cells in a time-dependent manner. At early stages of disease development, it enhances the ability of the SNS to polarize the Th response towards a more inflammatory state. Then, after disease is established, it enhances the ability of the SNS to dampen the inflammatory response via Tregs. The lack of concordance in results between global PACAP KO mice and mice with the PAC1 deletion targeted to catecholaminergic cells during early EAE may be explained by the fact that PACAP acts to regulate inflammation via multiple receptor subtypes and multiple targets, including inflammatory cells.


Asunto(s)
Células Cromafines/metabolismo , Neuronas Dopaminérgicas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología
10.
J Mol Neurosci ; 68(3): 452, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30362069

RESUMEN

The original version of this article unfortunately contained mistakes. The captured article title and corresponding author were incorrect.

11.
Neuro Oncol ; 20(6): 764-775, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29136244

RESUMEN

Background: Clinical trials of therapies directed against nodes of the signaling axis of phosphatidylinositol-3 kinase/Akt/mammalian target of rapamycin (mTOR) in glioblastoma (GBM) have had disappointing results. Resistance to mTOR inhibitors limits their efficacy. Methods: To determine mechanisms of resistance to chronic mTOR inhibition, we performed tandem screens on patient-derived GBM cultures. Results: An unbiased phosphoproteomic screen quantified phosphorylation changes associated with chronic exposure to the mTOR inhibitor rapamycin, and our analysis implicated a role for glycogen synthase kinase (GSK)3B attenuation in mediating resistance that was confirmed by functional studies. A targeted short hairpin RNA screen and further functional studies both in vitro and in vivo demonstrated that microtubule-associated protein (MAP)1B, previously associated predominantly with neurons, is a downstream effector of GSK3B-mediated resistance. Furthermore, we provide evidence that chronic rapamycin induces microtubule stability in a MAP1B-dependent manner in GBM cells. Additional experiments explicate a signaling pathway wherein combinatorial extracellular signal-regulated kinase (ERK)/mTOR targeting abrogates inhibitory phosphorylation of GSK3B, leads to phosphorylation of MAP1B, and confers sensitization. Conclusions: These data portray a compensatory molecular signaling network that imparts resistance to chronic mTOR inhibition in primary, human GBM cell cultures and points toward new therapeutic strategies.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/patología , Proteínas Asociadas a Microtúbulos/metabolismo , ARN Interferente Pequeño/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Comp Neurol ; 524(18): 3827-3848, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27197019

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP, gene name Adcyap1) regulates a wide variety of neurological and physiological functions, including metabolism and cognition, and plays roles in of multiple forms of stress. Because of its preferential expression in nerve fibers, it has often been difficult to trace and identify the endogenous sources of the peptide in specific populations of neurons. Here, we introduce a transgenic mouse line that harbors in its genome a bacterial artificial chromosome containing an enhanced green fluorescent protein (EGFP) expression cassette inserted upstream of the PACAP ATG translation initiation codon. Analysis of expression in brain sections of these mice using a GFP antibody reveals EGFP expression in distinct neuronal perikarya and dendritic arbors in several major brain regions previously reported to express PACAP from using a variety of approaches, including radioimmunoassay, in situ hybridization, and immunohistochemistry with and without colchicine. EGFP expression in neuronal perikarya was modulated in a manner similar to PACAP gene expression in motor neurons after peripheral axotomy in the ipsilateral facial motor nucleus in the brainstem, providing an example in which the transgene undergoes proper regulation in vivo. These mice and the high-resolution map obtained are expected to be useful in understanding the anatomical patterns of PACAP expression and its plasticity in the mouse. J. Comp. Neurol. 524:3827-3848, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Ratones Transgénicos , Modelos Animales , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Animales , Axotomía , Encéfalo/citología , Encéfalo/metabolismo , Traumatismos del Nervio Facial/metabolismo , Traumatismos del Nervio Facial/patología , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Masculino , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Médula Espinal/citología , Médula Espinal/metabolismo
13.
Psychopharmacology (Berl) ; 232(12): 2181-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25575489

RESUMEN

RATIONALE: An abundance of genetic and epidemiologic evidence as well as longitudinal neuroimaging data point to developmental origins for schizophrenia and other mental health disorders. Recent clinical studies indicate that microduplications of VIPR2, encoding the vasoactive intestinal peptide (VIP) receptor VPAC2, confer significant risk for schizophrenia and autism spectrum disorder. Lymphocytes from patients with these mutations exhibited higher VIPR2 gene expression and VIP responsiveness (cAMP induction), but mechanisms by which overactive VPAC2 signaling may lead to these psychiatric disorders are unknown. OBJECTIVES: We subcutaneously administered the highly selective VPAC2 receptor agonist Ro 25-1553 to C57BL/6 mice from postnatal day 1 (P1) to P14 to determine if overactivation of VPAC2 receptor signaling during postnatal brain maturation affects synaptogenesis and selected behaviors. RESULTS: Western blot analyses on P21 revealed significant reductions of synaptophysin and postsynaptic density protein 95 (PSD-95) in the prefrontal cortex, but not in the hippocampus in Ro 25-1553-treated mice. The same postnatally restricted treatment resulted in a disruption in prepulse inhibition of the acoustic startle measured in adult mice. No effects were observed in open-field locomotor activity, sociability in the three-chamber social interaction test, or fear conditioning or extinction. CONCLUSION: Overactivation of the VPAC2 receptor in the postnatal mouse results in a reduction in synaptic proteins in the prefrontal cortex and selective alterations in prepulse inhibition. These findings suggest that the VIPR2-linkage to mental health disorders may be due in part to overactive VPAC2 receptor signaling during a critical time of synaptic maturation.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Péptidos Cíclicos/farmacología , Inhibición Prepulso/efectos de los fármacos , Receptores de Tipo II del Péptido Intestinal Vasoactivo/agonistas , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Homólogo 4 de la Proteína Discs Large , Miedo/efectos de los fármacos , Femenino , Guanilato-Quinasas/metabolismo , Relaciones Interpersonales , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Embarazo , Reflejo de Sobresalto/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sinaptofisina/metabolismo , Péptido Intestinal Vasoactivo/farmacología
14.
J Comp Neurol ; 522(1): 169-85, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23818387

RESUMEN

Variants of the contactin associated protein-like 2 (Cntnap2) gene are risk factors for language-related disorders including autism spectrum disorder, specific language impairment, and stuttering. Songbirds are useful models for study of human speech disorders due to their shared capacity for vocal learning, which relies on similar cortico-basal ganglia circuitry and genetic factors. Here we investigate Cntnap2 protein expression in the brain of the zebra finch, a songbird species in which males, but not females, learn their courtship songs. We hypothesize that Cntnap2 has overlapping functions in vocal learning species, and expect to find protein expression in song-related areas of the zebra finch brain. We further expect that the distribution of this membrane-bound protein may not completely mirror its mRNA distribution due to the distinct subcellular localization of the two molecular species. We find that Cntnap2 protein is enriched in several song control regions relative to surrounding tissues, particularly within the adult male, but not female, robust nucleus of the arcopallium (RA), a cortical song control region analogous to human layer 5 primary motor cortex. The onset of this sexually dimorphic expression coincides with the onset of sensorimotor learning in developing males. Enrichment in male RA appears due to expression in projection neurons within the nucleus, as well as to additional expression in nerve terminals of cortical projections to RA from the lateral magnocellular nucleus of the nidopallium. Cntnap2 protein expression in zebra finch brain supports the hypothesis that this molecule affects neural connectivity critical for vocal learning across taxonomic classes.


Asunto(s)
Proteínas Aviares/metabolismo , Encéfalo/metabolismo , Pinzones/fisiología , Aprendizaje/fisiología , Proteínas del Tejido Nervioso/metabolismo , Vocalización Animal , Animales , Western Blotting , Encéfalo/crecimiento & desarrollo , Células Cultivadas , Femenino , Pinzones/crecimiento & desarrollo , Inmunohistoquímica , Masculino , Vías Nerviosas/metabolismo , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/metabolismo , Diferenciación Sexual
15.
Comp Cogn Behav Rev ; 9: 75-98, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26052371

RESUMEN

Language is a complex communicative behavior unique to humans, and its genetic basis is poorly understood. Genes associated with human speech and language disorders provide some insights, originating with the FOXP2 transcription factor, a mutation in which is the source of an inherited form of developmental verbal dyspraxia. Subsequently, targets of FOXP2 regulation have been associated with speech and language disorders, along with other genes. Here, we review these recent findings that implicate genetic factors in human speech. Due to the exclusivity of language to humans, no single animal model is sufficient to study the complete behavioral effects of these genes. Fortunately, some animals possess subcomponents of language. One such subcomponent is vocal learning, which though rare in the animal kingdom, is shared with songbirds. We therefore discuss how songbird studies have contributed to the current understanding of genetic factors that impact human speech, and support the continued use of this animal model for such studies in the future.

16.
J Neurophysiol ; 100(4): 2015-25, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18701760

RESUMEN

Cognitive and motor deficits associated with language and speech are seen in humans harboring FOXP2 mutations. The neural bases for FOXP2 mutation-related deficits are thought to reside in structural abnormalities distributed across systems important for language and motor learning including the cerebral cortex, basal ganglia, and cerebellum. In these brain regions, our prior research showed that FoxP2 mRNA expression patterns are strikingly similar between developing humans and songbirds. Within the songbird brain, this pattern persists throughout life and includes the striatal subregion, Area X, that is dedicated to song development and maintenance. The persistent mRNA expression suggests a role for FoxP2 that extends beyond the formation of vocal learning circuits to their ongoing use. Because FoxP2 is a transcription factor, a role in shaping circuits likely depends on FoxP2 protein levels which might not always parallel mRNA levels. Indeed our current study shows that FoxP2 protein, like its mRNA, is acutely downregulated in mature Area X when adult males sing with some differences. Total corticosterone levels associated with the different behavioral contexts did not vary, indicating that differences in FoxP2 levels are not likely attributable to stress. Our data, together with recent reports on FoxP2's target genes, suggest that lowered FoxP2 levels may allow for expression of genes important for circuit modification and thus vocal variability.


Asunto(s)
Pinzones/fisiología , Factores de Transcripción Forkhead/metabolismo , Neostriado/fisiología , Vocalización Animal/fisiología , Animales , Western Blotting , Química Encefálica/fisiología , Corticosterona/sangre , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Regulación hacia Abajo/fisiología , Técnica del Anticuerpo Fluorescente , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/fisiología , Humanos , Inmunohistoquímica , Neostriado/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Transcripción Genética
17.
J Neurophysiol ; 100(1): 108-16, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18480365

RESUMEN

Previous findings indicate that synaptic facilitation, a cellular mechanism underlying sensitization of the siphon withdrawal response (SWR) in Aplysia, depends on a cascade of postsynaptic events, including activation of inositol triphosphate (IP3) receptors and release of Ca2+ from postsynaptic intracellular stores. These findings suggest that phospholipase C (PLC), the enzyme that catalyzes IP3 formation, may play an important role in postsynaptic signaling during facilitation and learning in Aplysia. Using the PLC inhibitor U73122, we found that PLC activity is required for synaptic facilitation following a 10-min treatment with 5-HT, as measured at 20 min after 5-HT washout. Prior work has indicated that facilitation at this time is supported primarily by postsynaptic processes. To determine whether postsynaptic PLC activity is involved in 5-HT-mediated facilitatory actions, we examined the effect of U73122 on enhancement of the response of motor neurons isolated in cell culture to glutamate, the sensory neuron transmitter. A 10-min application of 5-HT induced persistent (>40 min) enhancement of glutamate-evoked potentials (Glu-EPs) recorded from isolated motor neurons, and this enhancement was blocked by U73122. Finally, we showed that injecting U73122 into intact animals before behavioral training impaired intermediate-term sensitization, indicating that PLC activity contributes to this form of nonassociative learning.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Neuronas Motoras/fisiología , Movimiento/fisiología , Neuronas Aferentes/fisiología , Sinapsis/fisiología , Fosfolipasas de Tipo C/metabolismo , Animales , Aplysia , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Cocultivo , Interacciones Farmacológicas , Estrenos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/farmacología , Movimiento/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Estimulación Física/métodos , Pirrolidinonas/farmacología , Serotonina/farmacología , Sinapsis/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...