Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38712156

RESUMEN

Summary: The stria vascularis (SV), part of the blood-labyrinth barrier, is an essential component of the inner ear that regulates the ionic environment required for hearing. SV degeneration disrupts cochlear homeostasis, leading to irreversible hearing loss, yet a comprehensive understanding of the SV, and consequently therapeutic availability for SV degeneration, is lacking. We developed a whole-tissue explant model from neonatal and adult mice to create a robust platform for SV research. We validated our model by demonstrating that the proliferative behaviour of the SV in vitro mimics SV in vivo, providing a representative model and advancing high-throughput SV research. We also provided evidence for pharmacological intervention in our system by investigating the role of Wnt/ß-catenin signaling in SV proliferation. Finally, we performed single-cell RNA sequencing from in vivo neonatal and adult mouse SV and revealed key genes and pathways that may play a role in SV proliferation and maintenance. Together, our results contribute new insights into investigating biological solutions for SV-associated hearing loss. Significance: Hearing loss impairs our ability to communicate with people and interact with our environment. This can lead to social isolation, depression, cognitive deficits, and dementia. Inner ear degeneration is a primary cause of hearing loss, and our study provides an in depth look at one of the major sites of inner ear degeneration: the stria vascularis. The stria vascularis and associated blood-labyrinth barrier maintain the functional integrity of the auditory system, yet it is relatively understudied. By developing a new in vitro model for the young and adult stria vascularis and using single cell RNA sequencing, our study provides a novel approach to studying this tissue, contributing new insights and widespread implications for auditory neuroscience and regenerative medicine. Highlights: - We established an organotypic explant system of the neonatal and adult stria vascularis with an intact blood-labyrinth barrier. - Proliferation of the stria vascularis decreases with age in vitro , modelling its proliferative behaviour in vivo . - Pharmacological studies using our in vitro SV model open possibilities for testing injury paradigms and therapeutic interventions. - Inhibition of Wnt signalling decreases proliferation in neonatal stria vascularis.- We identified key genes and transcription factors unique to developing and mature SV cell types using single cell RNA sequencing.

2.
Sci Rep ; 14(1): 3038, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321040

RESUMEN

The stria vascularis (SV) is a stratified epithelium in the lateral wall of the mammalian cochlea, responsible for both endolymphatic ion homeostasis and generation of the endocochlear potential (EP) critical for normal hearing. The SV has three layers consisting predominantly of basal, intermediate, and marginal cells. Intermediate and marginal cells form an intricate interdigitated network of cell projections making discrimination of the cells challenging. To enable intermediate cell visualization, we engineered by BAC transgenesis, reporter mouse lines expressing ZsGreen fluorescent protein under the control of Kcnj10 promoter and regulatory sequences. Kcnj10 encodes KCNJ10 protein (also known as Kir4.1 or Kir1.2), an ATP-sensitive inwardly-rectifying potassium channel critical to EP generation, highly expressed in SV intermediate cells. In these transgenic mice, ZsGreen fluorescence mimics Kcnj10 endogenous expression in the cochlea and was detected in the intermediate cells of the SV, in the inner phalangeal cells, Hensen's, Deiters' and pillar cells, in a subset of spiral ganglion neurons, and in glial cells. We show that expression of the transgene in hemizygous mice does not alter auditory function, nor EP. These transgenic Tg(Kcnj10-ZsGreen) mice allow live and fixed tissue visualization of ZsGreen-expressing intermediate cells and will facilitate future studies of stria vascularis cell function.


Asunto(s)
Oído Interno , Canales de Potasio de Rectificación Interna , Animales , Ratones , Estría Vascular/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Cóclea/metabolismo , Oído Interno/metabolismo , Ratones Transgénicos , Mamíferos/metabolismo
3.
Res Sq ; 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37886521

RESUMEN

The stria vascularis (SV) is a stratified epithelium in the lateral wall of the mammalian cochlea, responsible for both endolymphatic ion homeostasis and generation of the endocochlear potential (EP) critical for normal hearing. The SV has three layers consisting predominantly of basal, intermediate, and marginal cells. Intermediate and marginal cells form an intricate interdigitated network of cell projections making discrimination of the cells challenging. To enable intermediate cell visualization, we engineered by BAC transgenesis, reporter mouse lines expressing ZsGreen fluorescent protein under the control of Kcnj10 promoter and regulatory sequences. Kcnj10 encodes KCNJ10 protein (also known as Kir4.1 or Kir1.2), an ATP-sensitive inwardly-rectifying potassium channel critical to EP generation, highly expressed in SV intermediate cells. In these transgenic mice, ZsGreen fluorescence mimics Kcnj10 endogenous expression in the cochlea and was detected in the intermediate cells of the SV, in the inner phalangeal cells, Hensen's, Deiters' and pillar cells, in a subset of spiral ganglion neurons, and in glial cells. We show that expression of the transgene in hemizygous mice does not alter auditory function, nor EP These transgenic Tg(Kcnj10-ZsGreen) mice allow live and fixed tissue visualization of ZsGreen-expressing intermediate cells and will facilitate future studies of stria vascularis cell function.

4.
Front Mol Neurosci ; 14: 718241, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566577

RESUMEN

The endocochlear potential (EP) generated by the stria vascularis (SV) is necessary for hair cell mechanotransduction in the mammalian cochlea. We sought to create a model of EP dysfunction for the purposes of transcriptional analysis and treatment testing. By administering a single dose of cisplatin, a commonly prescribed cancer treatment drug with ototoxic side effects, to the adult mouse, we acutely disrupt EP generation. By combining these data with single cell RNA-sequencing findings, we identify transcriptional changes induced by cisplatin exposure, and by extension transcriptional changes accompanying EP reduction, in the major cell types of the SV. We use these data to identify gene regulatory networks unique to cisplatin treated SV, as well as the differentially expressed and druggable gene targets within those networks. Our results reconstruct transcriptional responses that occur in gene expression on the cellular level while identifying possible targets for interventions not only in cisplatin ototoxicity but also in EP dysfunction.

5.
Laryngoscope ; 129(1): E36-E40, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30325510

RESUMEN

Supporting cells (SCs) provide structure and maintain an environment that allows hair cells to receive and transmit signals in the auditory pathway. After insult to hair cells and ganglion cells, SCs respond by marking unsalvageable cells for death and maintain structural integrity. Although the histopathology after cochlear implantation has been described regarding hair cells and neural structures, surviving SCs in the implanted ear have not. We present a patient whose posthumous examination of an implanted cochlea demonstrated SC survival. This finding has implications for SC function in maintaining electrical hearing and candidacy for future hair cell regeneration therapies. Laryngoscope, 129:E36-E40, 2019.


Asunto(s)
Cóclea/citología , Implantación Coclear , Implantes Cocleares , Pérdida Auditiva Sensorineural/rehabilitación , Anciano de 80 o más Años , Supervivencia Celular , Humanos , Inmunohistoquímica , Masculino
6.
Sci Rep ; 8(1): 4403, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29535325

RESUMEN

Transmembrane proteins that mediate the cellular uptake or efflux of thyroid hormone potentially provide a key level of control over neurodevelopment. In humans, defects in one such protein, solute carrier SLC16A2 (MCT8) are associated with psychomotor retardation. Other proteins that transport the active form of thyroid hormone triiodothyronine (T3) or its precursor thyroxine (T4) have been identified in vitro but the wider significance of such transporters in vivo is unclear. The development of the auditory system requires thyroid hormone and the cochlea is a primary target tissue. We have proposed that the compartmental anatomy of the cochlea would necessitate transport mechanisms to convey blood-borne hormone to target tissues. We report hearing loss in mice with mutations in Slc16a2 and a related gene Slc16a10 (Mct10, Tat1). Deficiency of both transporters results in retarded development of the sensory epithelium similar to impairment caused by hypothyroidism, compounded with a progressive degeneration of cochlear hair cells and loss of endocochlear potential. Administration of T3 largely restores the development of the sensory epithelium and limited auditory function, indicating the T3-sensitivity of defects in the sensory epithelium. The results indicate a necessity for thyroid hormone transporters in cochlear development and function.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Sordera/genética , Eliminación de Gen , Células Ciliadas Auditivas/metabolismo , Proteínas de Transporte de Membrana/genética , Animales , Sordera/diagnóstico , Sordera/terapia , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Potenciales Evocados Auditivos , Femenino , Técnica del Anticuerpo Fluorescente , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células Ciliadas Auditivas/patología , Masculino , Ratones , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos , Órgano Espiral/metabolismo , Órgano Espiral/patología , Simportadores , Hormonas Tiroideas/sangre , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/farmacología
7.
Neurochem Res ; 42(9): 2646-2657, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28285415

RESUMEN

Glutamate carboxypeptidase II (GCPII) inactivates the peptide neurotransmitter N-acetylaspartylglutamate (NAAG) following synaptic release. Inhibitors of GCPII increase extracellular NAAG levels and are efficacious in animal models of clinical disorders via NAAG activation of a group II metabotropic glutamate receptor. mGluR2 and mGluR3 knock-out (ko) mice were used to test the hypothesis that mGluR3 mediates the activity of GCPII inhibitors ZJ43 and 2-PMPA in animal models of memory and memory loss. Short- (1.5 h) and long- (24 h) term novel object recognition tests were used to assess memory. Treatment with ZJ43 or 2-PMPA prior to acquisition trials increased long-term memory in mGluR2, but not mGluR3, ko mice. Nine month-old triple transgenic Alzheimer's disease model mice exhibited impaired short-term novel object recognition memory that was rescued by treatment with a NAAG peptidase inhibitor. NAAG peptidase inhibitors and the group II mGluR agonist, LY354740, reversed the short-term memory deficit induced by acute ethanol administration in wild type mice. 2-PMPA also moderated the effect of ethanol on short-term memory in mGluR2 ko mice but failed to do so in mGluR3 ko mice. LY354740 and ZJ43 blocked ethanol-induced motor activation. Both GCPII inhibitors and LY354740 also significantly moderated the loss of motor coordination induced by 2.1 g/kg ethanol treatment. These data support the conclusion that inhibitors of glutamate carboxypeptidase II are efficacious in object recognition models of normal memory and memory deficits via an mGluR3 mediated process, actions that could have widespread clinical applications.


Asunto(s)
Intoxicación Alcohólica/metabolismo , Enfermedad de Alzheimer/metabolismo , Glutamato Carboxipeptidasa II/metabolismo , Memoria/fisiología , Receptores de Glutamato Metabotrópico/deficiencia , Urea/análogos & derivados , Intoxicación Alcohólica/genética , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Antagonistas de Aminoácidos Excitadores/farmacología , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Glutamato Carboxipeptidasa II/genética , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Urea/farmacología
8.
Hum Mol Genet ; 23(18): 4960-9, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24847002

RESUMEN

The transactivation DNA-binding protein (TDP)-43 binds to thousands of mRNAs, but the functional outcomes of this binding remain largely unknown. TDP-43 binds to Park2 mRNA, which expresses the E3 ubiquitin ligase parkin. We previously demonstrated that parkin ubiquitinates TDP-43 and facilitates its translocation from the nucleus to the cytoplasm. Here we used brain penetrant tyrosine kinase inhibitors (TKIs), including nilotinib and bosutinib and showed that they reduce the level of nuclear TDP-43, abrogate its effects on neuronal loss, and reverse cognitive and motor decline. Nilotinib decreased soluble and insoluble TDP-43, while bosutinib did not affect the insoluble level. Parkin knockout mice exhibited high levels of endogenous TDP-43, while nilotinib and bosutinib did not alter TDP-43, underscoring an indispensable role for parkin in TDP-43 sub-cellular localization. These data demonstrate a novel functional relationship between parkin and TDP-43 and provide evidence that TKIs are potential therapeutic candidates for TDP-43 pathologies.


Asunto(s)
Cognición/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Destreza Motora/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Ubiquitina-Proteína Ligasas/metabolismo , Compuestos de Anilina/administración & dosificación , Compuestos de Anilina/farmacología , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Ratones , Ratones Transgénicos , Neuronas/patología , Nitrilos/administración & dosificación , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Quinolinas/administración & dosificación , Quinolinas/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
9.
Eur J Pharmacol ; 701(1-3): 27-32, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23200894

RESUMEN

The peptide neurotransmitter N-acetylaspartylglutamate (NAAG) is inactivated by the extracellular enzyme glutamate carboxypeptidase II. Inhibitors of this enzyme reverse dizocilpine (MK-801)-induced impairment of short-term memory in the novel object recognition test. The objective of this study was to test the hypothesis that NAAG peptidase inhibition enhances long-term (24h delay) memory of C57BL mice. These mice and mice in which glutamate carboxypeptidase II had been knocked out were presented with two identical objects to explore for 10min on day 1 and tested with one of these familiar objects and one novel object on day 2. Memory was assessed as the degree to which the mice recalled the familiar object and explored the novel object to a greater extent on day 2. Uninjected mice or mice injected with saline prior to the acquisition session on day 1 demonstrated a lack of memory of the acquisition experience by exploring the familiar and novel objects to the same extent on day 2. Mice treated with glutamate carboxypeptidase II inhibitors ZJ43 or 2-PMPA prior to the acquisition trial explored the novel object significantly more time than the familiar object on day 2. Consistent with these results, mice in which glutamate carboxypeptidase II had been knocked out distinguished the novel from the familiar object on day 2 while their heterozygous colony mates did not. Inhibition of glutamate carboxypeptidase II enhances recognition memory, a therapeutic action that might be useful in treatment of memory deficits related to age and neurological disorders.


Asunto(s)
Eliminación de Gen , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Glutamato Carboxipeptidasa II/genética , Memoria/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Animales , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Técnicas de Inactivación de Genes , Glutamato Carboxipeptidasa II/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos Organofosforados/farmacología , Urea/análogos & derivados , Urea/farmacología
10.
Eur J Neurosci ; 37(1): 118-29, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23134476

RESUMEN

Immunohistochemical studies previously revealed the presence of the peptide transmitter N-acetylaspartylglutamate (NAAG) in spinal motor neurons, axons and presumptive neuromuscular junctions (NMJs). At synapses in the central nervous system, NAAG has been shown to activate the type 3 metabotropic glutamate receptor (mGluR3) and is inactivated by an extracellular peptidase, glutamate carboxypeptidase II. The present study tested the hypothesis that NAAG meets the criteria for classification as a co-transmitter at the vertebrate NMJ. Confocal microscopy confirmed the presence of NAAG immunoreactivity and extended the resolution of the peptide's location in the lizard (Anolis carolinensis) NMJ. NAAG was localised to a presynaptic region immediately adjacent to postsynaptic acetylcholine receptors. NAAG was depleted by potassium-induced depolarisation and by electrical stimulation of motor axons. The NAAG receptor, mGluR3, was localised to the presynaptic terminal consistent with NAAG's demonstrated role as a regulator of synaptic release at central synapses. In contrast, glutamate receptors, type 2 metabotropic glutamate receptor (mGluR2) and N-methyl-d-aspartate, were closely associated with acetylcholine receptors in the postsynaptic membrane. Glutamate carboxypeptidase II, the NAAG-inactivating enzyme, was identified exclusively in perisynaptic glial cells. This localisation was confirmed by the loss of immunoreactivity when these cells were selectively eliminated. Finally, electrophysiological studies showed that exogenous NAAG inhibited evoked neurotransmitter release by activating a group II metabotropic glutamate receptor (mGluR2 or mGluR3). Collectively, these data support the conclusion that NAAG is a co-transmitter at the vertebrate NMJ.


Asunto(s)
Dipéptidos/farmacología , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Unión Neuromuscular/química , Neurotransmisores/farmacología , Animales , Dipéptidos/análisis , Agonistas de Aminoácidos Excitadores/farmacología , Glutamato Carboxipeptidasa II/análisis , Inmunohistoquímica , Lagartos , Neuronas Motoras/química , Neuronas Motoras/fisiología , N-Metilaspartato/farmacología , Unión Neuromuscular/fisiología , Potasio/farmacología , Terminales Presinápticos/química , Receptores Colinérgicos/análisis , Receptores de Glutamato Metabotrópico/análisis
11.
Mol Pain ; 8: 67, 2012 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-22971334

RESUMEN

BACKGROUND: Metabotropic glutamate receptors (mGluRs) have been identified as significant analgesic targets. Systemic treatments with inhibitors of the enzymes that inactivate the peptide transmitter N-acetylaspartylglutamate (NAAG), an mGluR3 agonist, have an analgesia-like effect in rat models of inflammatory and neuropathic pain. The goal of this study was to begin defining locations within the central pain pathway at which NAAG activation of its receptor mediates this effect. RESULTS: NAAG immunoreactivity was found in neurons in two brain regions that mediate nociceptive processing, the periaqueductal gray (PAG) and the rostral ventromedial medulla (RVM). Microinjection of the NAAG peptidase inhibitor ZJ43 into the PAG contralateral, but not ipsilateral, to the formalin injected footpad reduced the rapid and slow phases of the nociceptive response in a dose-dependent manner. ZJ43 injected into the RVM also reduced the rapid and slow phase of the response. The group II mGluR antagonist LY341495 blocked these effects of ZJ43 on the PAG and RVM. NAAG peptidase inhibition in the PAG and RVM did not affect the thermal withdrawal response in the hot plate test. Footpad inflammation also induced a significant increase in glutamate release in the PAG. Systemic injection of ZJ43 increased NAAG levels in the PAG and RVM and blocked the inflammation-induced increase in glutamate release in the PAG. CONCLUSION: These data demonstrate a behavioral and neurochemical role for NAAG in the PAG and RVM in regulating the spinal motor response to inflammation and that NAAG peptidase inhibition has potential as an approach to treating inflammatory pain via either the ascending (PAG) and/or the descending pain pathways (PAG and RVM) that warrants further study.


Asunto(s)
Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Inflamación/enzimología , Sustancia Gris Periacueductal/enzimología , Analgésicos/uso terapéutico , Animales , Formaldehído/toxicidad , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Bulbo Raquídeo/enzimología , Neuralgia/tratamiento farmacológico , Neuralgia/enzimología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo
12.
J Biol Chem ; 287(26): 21773-82, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22570482

RESUMEN

The "glutamate" theory of schizophrenia emerged from the observation that phencyclidine (PCP), an open channel antagonist of the NMDA subtype of glutamate receptor, induces schizophrenia-like behaviors in humans. PCP also induces a complex set of behaviors in animal models of this disorder. PCP also increases glutamate and dopamine release in the medial prefrontal cortex and nucleus accumbens, brain regions associated with expression of psychosis. Increased motor activation is among the PCP-induced behaviors that have been widely validated as models for the characterization of new antipsychotic drugs. The peptide transmitter N-acetylaspartylglutamate (NAAG) activates a group II metabotropic receptor, mGluR3. Polymorphisms in this receptor have been associated with schizophrenia. Inhibitors of glutamate carboxypeptidase II, an enzyme that inactivates NAAG following synaptic release, reduce several behaviors induced by PCP in animal models. This research tested the hypothesis that two structurally distinct NAAG peptidase inhibitors, ZJ43 and 2-(phosphonomethyl)pentane-1,5-dioic acid, would elevate levels of synaptically released NAAG and reduce PCP-induced increases in glutamate and dopamine levels in the medial prefrontal cortex and nucleus accumbens. NAAG-like immunoreactivity was found in neurons and presumptive synaptic endings in both regions. These peptidase inhibitors reduced the motor activation effects of PCP while elevating extracellular NAAG levels. They also blocked PCP-induced increases in glutamate but not dopamine or its metabolites. The mGluR2/3 antagonist LY341495 blocked these behavioral and neurochemical effects of the peptidase inhibitors. The data reported here provide a foundation for assessment of the neurochemical mechanism through which NAAG achieves its antipsychotic-like behavioral effects and support the conclusion NAAG peptidase inhibitors warrant further study as a novel antipsychotic therapy aimed at mGluR3.


Asunto(s)
Dopamina/química , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Ácido Glutámico/química , Núcleo Accumbens/metabolismo , Fenciclidina/farmacología , Corteza Prefrontal/metabolismo , Esquizofrenia/fisiopatología , Animales , Antipsicóticos/farmacología , Conducta Animal , Modelos Animales de Enfermedad , Dopamina/metabolismo , Glutamato Carboxipeptidasa II/química , Ácido Glutámico/metabolismo , Masculino , Neurotransmisores/metabolismo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/tratamiento farmacológico
14.
J Neurochem ; 118(4): 490-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21644997

RESUMEN

A substantial body of data was reported between 1984 and 2000 demonstrating that the neuropeptide N-acetylaspartylglutamate (NAAG) not only functions as a neurotransmitter but also is the third most prevalent transmitter in the mammalian nervous system behind glutamate and GABA. By 2005, this conclusion was validated further through a series of studies in vivo and in vitro. The primary enzyme responsible for the inactivation of NAAG following its synaptic release had been cloned, characterized and knocked out. Potent inhibitors of this enzyme were developed and their efficacy has been extensively studied in a series of animal models of clinical conditions, including stroke, peripheral neuropathy, traumatic brain injury, inflammatory and neuropathic pain, cocaine addiction, and schizophrenia. Considerable progress also has been made in defining further the mechanism of action of these peptidase inhibitors in elevating synaptic levels of NAAG with the consequent inhibition of transmitter release via the activation of pre-synaptic metabotropic glutamate receptor 3 by this peptide. Very recent discoveries include identification of two different nervous system enzymes that mediate the synthesis of NAAG from N-acetylaspartate and glutamate and the finding that one of these enzymes also mediates the synthesis of a second member of the NAAG family of neuropeptides, N-acetylaspartylglutamylglutamate.


Asunto(s)
Dipéptidos/fisiología , Neuropéptidos/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Lesiones Encefálicas/tratamiento farmacológico , Dipéptidos/genética , Dipéptidos/metabolismo , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Humanos , Hiperalgesia/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Neuropéptidos/genética , Neuropéptidos/metabolismo , Neurotransmisores/fisiología , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Esquizofrenia/tratamiento farmacológico , Trastornos Relacionados con Sustancias/terapia
15.
Brain Res ; 1395: 62-73, 2011 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565332

RESUMEN

Traumatic brain injury (TBI) leads to a rapid and excessive increase in glutamate concentration in the extracellular milieu, which is strongly associated with excitotoxicity and neuronal degeneration. N-acetylaspartylglutamate (NAAG), a prevalent peptide neurotransmitter in the vertebrate nervous system, is released along with glutamate and suppresses glutamate release by actions at pre-synaptic metabotropic glutamate autoreceptors. Extracellular NAAG is hydrolyzed to N-acetylaspartate and glutamate by peptidase activity. In the present study PGI-02776, a newly designed di-ester prodrug of the urea-based NAAG peptidase inhibitor ZJ-43, was tested for neuroprotective potential when administered intraperitoneally 30 min after lateral fluid percussion TBI in the rat. Stereological quantification of hippocampal CA2-3 degenerating neurons at 24 h post injury revealed that 10 mg/kg PGI-02776 significantly decreased the number of degenerating neurons (p<0.05). Both average latency analysis of Morris water maze performance and assessment of 24-hour memory retention revealed significant differences between sham-TBI and TBI-saline. In contrast, no significant difference was found between sham-TBI and PGI-02776 treated groups in either analysis indicating an improvement in cognitive performance with PGI-02776 treatment. Histological analysis on day 16 post-injury revealed significant cell death in injured animals regardless of treatment. In vitro NAAG peptidase inhibition studies demonstrated that the parent compound (ZJ-43) exhibited potent inhibitory activity while the mono-ester (PGI-02749) and di-ester (PGI-02776) prodrug compounds exhibited moderate and weak levels of inhibitory activity, respectively. Pharmacokinetic assays in uninjured animals found that the di-ester (PGI-02776) crossed the blood-brain barrier. PGI-02776 was also readily hydrolyzed to both the mono-ester (PGI-02749) and the parent compound (ZJ-43) in both blood and brain. Overall, these findings suggest that post-injury treatment with the ZJ-43 prodrug PGI-02776 reduces both acute neuronal pathology and longer term cognitive deficits associated with TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Inhibidores de Proteasas/farmacocinética , Urea/análogos & derivados , Animales , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Glutamato Carboxipeptidasa II/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/aislamiento & purificación , Neurotransmisores/metabolismo , Neurotransmisores/fisiología , Inhibidores de Proteasas/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Urea/aislamiento & purificación , Urea/farmacología
16.
Psychopharmacology (Berl) ; 216(2): 235-43, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21327758

RESUMEN

RATIONALE: Group II metabotropic glutamate receptor (mGluR) agonists represent a novel approach to the treatment of schizophrenia. Inasmuch as the peptide neurotransmitter N-acetylaspartylglutamate (NAAG) activates these receptors, NAAG peptidase inhibitors conceptually represent a parallel path toward development of new antipsychotic drugs. While group II agonists are effective in several animal models of schizophrenia, they are reported to lack efficacy in moderating the effects of phencyclidine (PCP) on prepulse inhibition of acoustic startle in animal models of sensory processing deficits found in this disorder. OBJECTIVE: The objective of this study was to re-examine the efficacy of a group II metabotropic glutamate agonist and NAAG peptidase inhibitors in prepulse inhibition models of schizophrenia across two strains of mice. METHODS: The method used was an assay to determine the efficacy of these drugs in moderating the reduction in prepulse inhibition of acoustic startle in mice treated with PCP and D: -amphetamine. RESULTS: The group II agonist LY354740 (5 and 10 mg/kg) moderated the effects of PCP on prepulse inhibition of acoustic startle in DBA/2 but not C57BL/6 mice. In contrast, two NAAG peptidase inhibitors, ZJ43 (150 mg/kg) and 2-PMPA (50, 100, and 150 mg/kg), did not significantly affect the PCP-induced reduction in prepulse inhibition in either strain. CONCLUSIONS: These data demonstrate that the efficacy of group II agonists in this model of sensory motor processing is strain-specific in mice. The difference between the effects of the group II agonist and the peptidase inhibitors in the DBA/2 mice may relate to the difference in efficacy of NAAG and the agonist at mGluR2.


Asunto(s)
Compuestos Bicíclicos con Puentes/farmacología , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/agonistas , Esquizofrenia/tratamiento farmacológico , Animales , Compuestos Bicíclicos con Puentes/administración & dosificación , Dextroanfetamina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/administración & dosificación , Agonistas de Aminoácidos Excitadores/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/farmacología , Fenciclidina , Reflejo de Sobresalto/efectos de los fármacos , Esquizofrenia/fisiopatología , Especificidad de la Especie , Urea/análogos & derivados , Urea/farmacología
17.
Biol Psychiatry ; 63(1): 86-91, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17597589

RESUMEN

BACKGROUND: N-methyl-D-aspartate (NMDA) receptor open channel blockers phencyclidine (PCP) and dizocilpine (MK-801) elicit schizophrenia-like symptoms in humans and in animal models. Group II metabotropic glutamate receptor agonists reverse the behavioral effects of PCP and MK-801 in animal models. N-acetylaspartylglutamate (NAAG), the third most prevalent neurotransmitter in the mammalian nervous system, is a selective group II metabotropic glutamate receptor agonist. We previously reported that ZJ43, a potent inhibitor of the enzymes that inactivate synaptically released NAAG, reduced motor and stereotypic effects of PCP in the rat. METHODS: To confirm the efficacy of NAAG peptidase inhibition in decreasing motor behaviors induced by PCP and MK-801, ZJ43 was tested in additional schizophrenia models. RESULTS: ZJ43 reduced MK-801-induced motor activation in a mouse model that has been used to characterize the efficacy of a wide range of pharmacotherapies for this human disorder. In a second mouse strain, the peptidase inhibitor reduced PCP-induced stereotypic movements. ZJ43 also reduced PCP-induced negative symptoms in a resident-intruder assay. The group II metabotropic glutamate receptor antagonist, LY341495, blocked the effect of NAAG peptidase inhibition in these mouse models of positive and negative PCP- and MK-801-induced behaviors. Additionally, LY341495 alone increased some PCP-induced behaviors suggesting that normal levels of NAAG act to moderate the effect of PCP via a group II mGluR. CONCLUSIONS: These data support the proposal that NAAG peptidase inhibition and elevation of synaptic NAAG levels represent a new therapeutic approach to treating the positive and negative symptoms of schizophrenia that are modeled by open channel NMDA receptor antagonists.


Asunto(s)
Conducta Animal/efectos de los fármacos , Maleato de Dizocilpina , Glutamato Carboxipeptidasa II/uso terapéutico , Fenciclidina , Receptores de Glutamato Metabotrópico/fisiología , Esquizofrenia , Conducta Agonística/efectos de los fármacos , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Conducta Exploratoria/efectos de los fármacos , Masculino , Ratones , Esquizofrenia/inducido químicamente , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología , Conducta Estereotipada/efectos de los fármacos , Urea/análogos & derivados , Urea/farmacología
18.
J Neurochem ; 96(4): 1071-7, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16417588

RESUMEN

Metabotropic receptors may couple to different G proteins in different cells or perhaps even in different regions of the same cell. To date, direct studies of group II and group III metabotropic glutamate receptors' (mGluRs) relationships to second messenger cascades have reported negative coupling of these receptors to cyclic AMP (cAMP) levels in neurons, astrocytes and transfected cells. In the present study, we found that the peptide neurotransmitter N-acetylaspartylglutamate (NAAG), an mGluR3-selective agonist, decreased sodium nitroprusside (SNP)-stimulated cyclic GMP (cGMP) levels in cerebellar granule cells and cerebellar astrocytes. The mGluR3 and group II agonists FN6 and LY354740 had similar effects on cGMP levels. The mGluR3 and group II antagonists beta-NAAG and LY341495 blocked these actions. Treatment with pertussis toxin inhibited the effects of NAAG on SNP-stimulated cGMP levels in rat cerebellar astrocytes but not in cerebellar neurons. These data support the conclusion that mGluR3 is also coupled to cGMP levels and that this mGluR3-induced reduction of cGMP levels is mediated by different G proteins in cerebellar astrocytes and neurons. We previously reported that this receptor is coupled to a cAMP cascade via a pertussis toxin-sensitive G protein in cerebellar neurons, astrocytes and transfected cells. Taken together with the present data, we propose that mGluR3 is coupled to two different G proteins in granule cell neurons. These data greatly expand knowledge of the range of second messenger cascades induced by mGluR3, and have implications for clinical conditions affected by NAAG and other group II mGluR agonists.


Asunto(s)
Astrocitos/fisiología , GMP Cíclico/metabolismo , Neuronas/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Aminoácidos/farmacología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Cerebelo/citología , Cerebelo/fisiología , Dipéptidos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Proteínas de Unión al GTP/metabolismo , Neuronas/efectos de los fármacos , Nitroprusiato/farmacología , Toxina del Pertussis/farmacología , Ratas , Ratas Sprague-Dawley , Xantenos/farmacología
19.
Trends Pharmacol Sci ; 26(9): 477-84, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16055199

RESUMEN

N-Acetylaspartylglutamate (NAAG) is the most abundant and widely distributed peptide transmitter in the mammalian nervous system. NAAG activates the metabotropic glutamate mGlu(3) receptor at presynaptic sites, inhibiting the release of neurotransmitters, including glutamate, and activates mGlu(3) receptors on glial cells, stimulating the release of neuroprotective growth factors from these cells. Elevated levels of glutamate released from neurons are associated with the pathology of stroke, traumatic nervous system injury, amyotrophic lateral sclerosis, inflammatory and neuropathic pain, diabetic neuropathy and the schizophrenia-like symptoms elicited by phencyclidine. NAAG is inactivated by specific peptidases following its synaptic release. Novel compounds that inhibit these enzymes prolong the activity of synaptically released NAAG and have significant therapeutic efficacy in animal models of these diverse clinical conditions. In this review, we summarize recent studies in these animal models and discuss the mechanisms by which NAAG peptidase inhibitors achieve these effects.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Dipéptidos/metabolismo , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Esquizofrenia/tratamiento farmacológico , Animales , Humanos
20.
Clin Cancer Res ; 11(11): 4022-8, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930336

RESUMEN

PURPOSE: Prostate-specific membrane antigen (PSMA) is a cell surface protein that is overexpressed in prostate cancer, including hormone-refractory and metastatic disease. Our goal in this study was to develop a series of PSMA-based imaging agents for clinical use. EXPERIMENTAL DESIGN: We have synthesized and evaluated the in vivo biodistribution of two radiolabeled urea derivatives that have high affinity for PSMA in severe combined immunodeficient mice harboring MCF-7 (breast, PSMA-negative), PC-3 (prostate, PSMA-negative), and LNCaP (prostate, PSMA-positive) xenografts. Radiopharmaceutical binding selectivity and tumor uptake were also evaluated in vivo using dedicated small animal positron emission tomography, single photon emission computed tomography, and gamma scintigraphic imaging devices. N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-S-[(11)C]methyl-L-cysteine ([(11)C]DCMC K(i), 3.1 nmol/L) and N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-S-3-[(125)I]iodo-L-tyrosine ([(125)C]DCIT K(i), 1.5 nmol/L) were synthesized using [(11)C]CH(3)I and with [(125)I]NaI/Iodogen, respectively. RESULTS: At 30 minutes postinjection, [(11)C]DCMC and [(125)I]DCIT showed tumor/muscle ratios of 10.8 and 4.7, respectively, with clear delineation of LNCaP-derived tumors on imaging. MCF-7- and PC-3-derived tumors showed significantly less uptake of [(11)C]DCMC or [(125)I]DCIT. CONCLUSION: These results show the feasibility of imaging PSMA-positive prostate cancer using low molecular weight agents.


Asunto(s)
Antígenos de Superficie/metabolismo , Glutamato Carboxipeptidasa II/metabolismo , Tomografía de Emisión de Positrones/métodos , Neoplasias de la Próstata/diagnóstico por imagen , Animales , Unión Competitiva , Radioisótopos de Carbono , Línea Celular Tumoral , Femenino , Humanos , Radioisótopos de Yodo , Ligandos , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Radiofármacos/síntesis química , Radiofármacos/metabolismo , Radiofármacos/farmacocinética , Distribución Tisular , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...