Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biomed Sci ; 28(1): 70, 2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34635103

RESUMEN

In modern societies, with an increase in the older population, age-related neurodegenerative diseases have progressively become greater socioeconomic burdens. To date, despite the tremendous effort devoted to understanding neurodegenerative diseases in recent decades, treatment to delay disease progression is largely ineffective and is in urgent demand. The development of new strategies targeting these pathological features is a timely topic. It is important to note that most degenerative diseases are associated with the accumulation of specific misfolded proteins, which is facilitated by several common features of neurodegenerative diseases (including poor energy homeostasis and mitochondrial dysfunction). Adenosine is a purine nucleoside and neuromodulator in the brain. It is also an essential component of energy production pathways, cellular metabolism, and gene regulation in brain cells. The levels of intracellular and extracellular adenosine are thus tightly controlled by a handful of proteins (including adenosine metabolic enzymes and transporters) to maintain proper adenosine homeostasis. Notably, disruption of adenosine homeostasis in the brain under various pathophysiological conditions has been documented. In the past two decades, adenosine receptors (particularly A1 and A2A adenosine receptors) have been actively investigated as important drug targets in major degenerative diseases. Unfortunately, except for an A2A antagonist (istradefylline) administered as an adjuvant treatment with levodopa for Parkinson's disease, no effective drug based on adenosine receptors has been developed for neurodegenerative diseases. In this review, we summarize the emerging findings on proteins involved in the control of adenosine homeostasis in the brain and discuss the challenges and future prospects for the development of new therapeutic treatments for neurodegenerative diseases and their associated disorders based on the understanding of adenosine homeostasis.


Asunto(s)
Adenosina/fisiología , Encéfalo/fisiopatología , Homeostasis , Enfermedades Neurodegenerativas/fisiopatología , Proteínas/metabolismo , Humanos
2.
J Neurosci ; 39(8): 1505-1524, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30587542

RESUMEN

Intrinsically photosensitive retinal ganglion cells (ipRGCs), which express the photopigment melanopsin, are photosensitive neurons in the retina and are essential for non-image-forming functions, circadian photoentrainment, and pupillary light reflexes. Five subtypes of ipRGCs (M1-M5) have been identified in mice. Although ipRGCs are spared in several forms of inherited blindness, they are affected in Alzheimer's disease and aging, which are associated with impaired circadian rhythms. Huntington's disease (HD) is an autosomal neurodegenerative disease caused by the expansion of a CAG repeat in the huntingtin gene. In addition to motor function impairment, HD mice also show impaired circadian rhythms and loss of ipRGC. Here, we found that, in HD mouse models (R6/2 and N171-82Q male mice), the expression of melanopsin was reduced before the onset of motor deficits. The expression of retinal T-box brain 2, a transcription factor essential for ipRGCs, was associated with the survival of ipRGCs. The number of M1 ipRGCs in R6/2 male mice was reduced due to apoptosis, whereas non-M1 ipRGCs were relatively resilient to HD progression. Most importantly, the reduced innervations of M1 ipRGCs, which was assessed by X-gal staining in R6/2-OPN4Lacz/+ male mice, contributed to the diminished light-induced c-fos and vasoactive intestinal peptide in the suprachiasmatic nuclei (SCN), which may explain the impaired circadian photoentrainment in HD mice. Collectively, our results show that M1 ipRGCs were susceptible to the toxicity caused by mutant Huntingtin. The resultant impairment of M1 ipRGCs contributed to the early degeneration of the ipRGC-SCN pathway and disrupted circadian regulation during HD progression.SIGNIFICANCE STATEMENT Circadian disruption is a common nonmotor symptom of Huntington's disease (HD). In addition to the molecular defects in the suprachiasmatic nuclei (SCN), the cause of circadian disruption in HD remains to be further explored. We hypothesized that ipRGCs, by integrating light input to the SCN, participate in the circadian regulation in HD mice. We report early reductions in melanopsin in two mouse models of HD, R6/2, and N171-82Q. Suppression of retinal T-box brain 2, a transcription factor essential for ipRGCs, by mutant Huntingtin might mediate the reduced number of ipRGCs. Importantly, M1 ipRGCs showed higher susceptibility than non-M1 ipRGCs in R6/2 mice. The resultant impairment of M1 ipRGCs contributed to the early degeneration of the ipRGC-SCN pathway and the circadian abnormality during HD progression.


Asunto(s)
Ritmo Circadiano/fisiología , Enfermedad de Huntington/patología , Células Ganglionares de la Retina/patología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas del Ojo/biosíntesis , Genes Reporteros , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Actividad Motora , Reflejo Anormal , Reflejo Pupilar , Células Ganglionares de la Retina/efectos de la radiación , Opsinas de Bastones/biosíntesis , Núcleo Supraquiasmático/metabolismo , Proteínas de Dominio T Box/biosíntesis , Péptido Intestinal Vasoactivo/biosíntesis
3.
Mov Disord ; 32(11): 1600-1609, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28782830

RESUMEN

BACKGROUND: Disruptions in gamma-aminobutyric (GABA) acid signaling are believed to be involved in Huntington's disease pathogenesis, but the regulation of GABAergic signaling remains elusive. Here we evaluated GABAergic signaling by examining the function of GABAergic drugs in Huntington's disease and the expression of GABAergic molecules using mouse models and human brain tissues from Huntington's disease. METHODS: We treated wild-type and R6/2 mice (a transgenic Huntington's disease mouse model) acutely with vehicle, diazepam, or gaboxadol (drugs that selectively target synaptic or extrasynaptic GABAA receptors) and monitored their locomotor activity. The expression levels of GABAA receptors and a major neuron-specific chloride extruder (potassium-chloride cotransporter-2) were analyzed by real-time quantitative polymerase chain reaction, Western blot, and immunocytochemistry. RESULTS: The R6/2 mice were less sensitive to the sedative effects of both drugs, suggesting reduced function of GABAA receptors. Consistently, the expression levels of α1/α2 and δ subunits were lower in the cortex and striatum of R6/2 mice. Similar results were also found in 2 other mouse models of Huntington's disease and in Huntington's disease patients. Moreover, the interaction and expression levels of potassium-chloride cotransporter-2 and its activator (brain-type creatine kinase) were decreased in Huntington's disease neurons. These findings collectively suggest impaired chloride homeostasis, which further dampens GABAA receptor-mediated inhibitory signaling in Huntington's disease brains. CONCLUSIONS: The dysregulated GABAergic responses and altered expression levels of GABAA receptors and potassium-chloride cotransporter-2 in Huntington's disease mice appear to be authentic and may contribute to the clinical manifestations of Huntington's disease patients. © 2017 International Parkinson and Movement Disorder Society.


Asunto(s)
Conducta Animal/efectos de los fármacos , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Diazepam/farmacología , Agonistas del GABA/farmacología , Moduladores del GABA/farmacología , Enfermedad de Huntington/metabolismo , Isoxazoles/farmacología , Receptores de GABA-A/metabolismo , Animales , Corteza Cerebral/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Receptores de GABA-A/efectos de los fármacos
4.
Neurobiol Dis ; 96: 47-53, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27567601

RESUMEN

The initial goal of this study was to investigate alterations in adenosine A2A receptor (A2AR) density or function in a rat model of Huntington disease (HD) with reported insensitivity to an A2AR antagonist. Unsuspected negative results led to the hypothesis of a low striatal adenosine tone and to the search for the mechanisms involved. Extracellular striatal concentrations of adenosine were measured with in vivo microdialysis in two rodent models of early neuropathological stages of HD disease, the Tg51 rat and the zQ175 knock-in mouse. In view of the crucial role of the equilibrative nucleoside transporter (ENT1) in determining extracellular content of adenosine, the binding properties of the ENT1 inhibitor [3H]-S-(4-Nitrobenzyl)-6-thioinosine were evaluated in zQ175 mice and the differential expression and differential coexpression patterns of the ENT1 gene (SLC29A1) were analyzed in a large human cohort of HD disease and controls. Extracellular striatal levels of adenosine were significantly lower in both animal models as compared with control littermates and striatal ENT1 binding sites were significantly upregulated in zQ175 mice. ENT1 transcript was significantly upregulated in HD disease patients at an early neuropathological severity stage, but not those with a higher severity stage, relative to non-demented controls. ENT1 transcript was differentially coexpressed (gained correlations) with several other genes in HD disease subjects compared to the control group. The present study demonstrates that ENT1 and adenosine constitute biomarkers of the initial stages of neurodegeneration in HD disease and also predicts that ENT1 could constitute a new therapeutic target to delay the progression of the disease.


Asunto(s)
Biomarcadores/metabolismo , Cuerpo Estriado/metabolismo , Regulación de la Expresión Génica/genética , Enfermedad de Huntington/patología , Proteínas de Transporte de Nucleósidos/metabolismo , Corteza Prefrontal/metabolismo , Adenosina/metabolismo , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/complicaciones , Enfermedad de Huntington/genética , Locomoción/genética , Trastornos Psicomotores/tratamiento farmacológico , Trastornos Psicomotores/etiología , Purinas/uso terapéutico , Ratas , Ratas Transgénicas , Receptor de Adenosina A2A/metabolismo , Triazinas/farmacocinética , Triazoles/farmacocinética , Expansión de Repetición de Trinucleótido/genética , Tritio/farmacocinética
5.
Biochim Biophys Acta ; 1842(9): 1668-80, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24946181

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurological disorder that is induced by a CAG trinucleotide expansion in exon 1 of the Huntingtin (HTT) gene. We previously reported that the abnormal activation of an important energy sensor, AMP-activated protein kinase α1 (AMPK-α1), occurs in the brains of mice and patients with HD, which suggests that this abnormal activation may contribute to neuronal degeneration in HD. In the present study, we demonstrated that the elevated oxidative stress that was evoked by a polyQ-expanded mutant HTT (mHTT) caused the abnormal activation of AMPK-α1 and, subsequently, resulted in neurotoxicity in a striatal progenitor cell line (STHdh(Q109)) and in the striatum of a transgenic mouse model of HD (R6/2). The systematic administration of an antioxidant (N-acetyl-cysteine, NAC) to R6/2 mice suppressed the activation of AMPK-α1, reduced neuronal toxicity, which was assessed by the activation of caspases, increased neuronal density, ameliorated ventricle enlargement, and improved motor dysfunction. This beneficial effect of NAC in vivo appears to be direct because NAC also reduced the activation of AMPK-α1 and the death of STHdh(Q109) cells upon elevated oxidative stress. Moreover, the activation of AMPK enhanced the level of oxidative stress in STHdh(Q109) cells, in primary neurons of R6/2 mice, and in the striatum of two different HD mouse models (R6/2 and Hdh(150Q/+)), whereas the inhibition of AMPK reduced the level of oxidative stress. Collectively, our findings suggest that positive feedback regulation between the elevated oxidative stress and the activation of AMPK-α1 contributes to the progression of HD.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Atrofia/patología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/fisiología , Neuronas/patología , Estrés Oxidativo , Animales , Apoptosis , Atrofia/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Cuerpo Estriado/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Transgénicos , Degeneración Nerviosa , Neuronas/metabolismo
6.
Hum Mol Genet ; 19(20): 4043-58, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20668093

RESUMEN

Huntington's disease (HD) is a neurodegenerative disease caused by the expansion of a CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene. Here, we report that the transcript of the peroxisome proliferator-activated receptor-γ (PPARγ), a transcription factor that is critical for energy homeostasis, was markedly downregulated in multiple tissues of a mouse model (R6/2) of HD and in lymphocytes of HD patients. Therefore, downregulation of PPARγ seems to be a pathomechanism of HD. Chronic treatment of R6/2 mice with an agonist of PPARγ (thiazolidinedione, TZD) rescued progressive weight loss, motor deterioration, formation of mutant Htt aggregates, jeopardized global ubiquitination profiles, reduced expression of two neuroprotective proteins (brain-derived neurotrophic factor and Bcl-2) and shortened life span exhibited by these mice. By reducing HTT aggregates and, thus, ameliorating the recruitment of PPARγ into HTT aggregates, chronic TZD treatment also elevated the availability of the PPARγ protein and subsequently normalized the expression of two of its downstream genes (the glucose transporter type 4 and PPARγ coactivator-1 alpha genes). The protective effects described above appear to have been exerted, at least partially, via direct activation of PPARγ in the brain, as TZD was detected in the brains of mice treated with TZD and because a PPARγ agonist (rosiglitazone) protected striatal cells from mHTT-evoked energy deficiency and toxicity. We demonstrated that the systematic downregulation of PPARγ seems to play a critical role in the dysregulation of energy homeostasis observed in HD, and that PPARγ is a potential therapeutic target for this disease.


Asunto(s)
Metabolismo Energético , Enfermedad de Huntington/metabolismo , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Adipocitos/metabolismo , Animales , Encéfalo/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Hígado/metabolismo , Linfocitos/metabolismo , Ratones , PPAR gamma/agonistas , PPAR gamma/deficiencia , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Reacción en Cadena de la Polimerasa , Rosiglitazona , Tiazolidinedionas/administración & dosificación , Transactivadores/genética , Factores de Transcripción
7.
Acta Neuropathol Commun ; 9(1): 112, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158119

RESUMEN

Tau pathology is instrumental in the gradual loss of neuronal functions and cognitive decline in tauopathies, including Alzheimer's disease (AD). Earlier reports showed that adenosine metabolism is abnormal in the brain of AD patients while consequences remained ill-defined. Herein, we aimed at investigating whether manipulation of adenosine tone would impact Tau pathology, associated molecular alterations and subsequent neurodegeneration. We demonstrated that treatment with an inhibitor (J4) of equilibrative nucleoside transporter 1 (ENT1) exerted beneficial effects in a mouse model of Tauopathy. Treatment with J4 not only reduced Tau hyperphosphorylation but also rescued memory deficits, mitochondrial dysfunction, synaptic loss, and abnormal expression of immune-related gene signatures. These beneficial effects were particularly ascribed to the ability of J4 to suppress the overactivation of AMPK (an energy reduction sensor), suggesting that normalization of energy dysfunction mitigates neuronal dysfunctions in Tauopathy. Collectively, these data highlight that targeting adenosine metabolism is a novel strategy for tauopathies.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/patología , Tranportador Equilibrativo 1 de Nucleósido/antagonistas & inhibidores , Tauopatías/metabolismo , Tauopatías/patología , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones
8.
J Cell Physiol ; 223(3): 679-86, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20143335

RESUMEN

Glucocorticoids (GCs) have been employed as immunosuppressive agents for many years. However, it is still unclear how GCs instantly uncouple T cells from acute stressful inflammatory. In terms of time scale, the genomic activity of the classic GC receptor cannot fulfill this role under crisis; but a rapid non-genomic response can. In a previous study, intracellular acidification was found to be due to a rapid non-genomic inhibition of Na(+)/H(+)-exchange 1 (NHE1) and this event led to the immunosuppression of T cell proliferation by progesterone. The aim of this study was to examine whether there is a rapid acidification response caused by an inhibition of NHE1 activity and to explore the differential non-genomic effect on immunosuppression of hydrocortisone and dexamethasone. The IC(50) values for NHE1-dependent pH(i) recovery by hydrocortisone and dexamethasone are 250 and 1 nM, respectively. Co-stimulation of GCs with phytohemagglutinin (PHA) is able to inhibit PHA-induced IL-2 secretion, IL-4 secretion, and T-cell proliferation. Furthermore, apoptosis in PHA-activated T cells is not induced by hydrocortisone but by dexamethasone. The mechanism of immunosuppression on proliferation by dexamethasone was found to be different of hydrocortisone and seems to involve cytotoxicity against T cells. Moreover, apoptosis induced by dexamethasone and impermeable dexamethasone-bovine serum albumin suggests that the apoptotic immunosuppression occurs through both the plasma membrane and cytoplasmic sites. The rapid inhibitory responses triggered by GCs would seem to release T cells instantly when an acute stress-related response is needed. Nonetheless, the apoptotic immunosuppression by dexamethasone is attributable to its severe cytotoxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Transporte de Catión/antagonistas & inhibidores , Dexametasona/farmacología , Glucocorticoides/farmacología , Hidrocortisona/farmacología , Terapia de Inmunosupresión , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Linfocitos T/inmunología , Adulto , Amilorida/análogos & derivados , Amilorida/farmacología , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Genoma Humano/genética , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Intercambiador 1 de Sodio-Hidrógeno , Linfocitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Timidina/metabolismo , Tritio , Adulto Joven
9.
Nat Commun ; 10(1): 3473, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31375685

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder that manifests with movement dysfunction. The expression of mutant Huntingtin (mHTT) disrupts the functions of brain cells. Galectin-3 (Gal3) is a lectin that has not been extensively explored in brain diseases. Herein, we showed that the plasma Gal3 levels of HD patients and mice correlated with disease severity. Moreover, brain Gal3 levels were higher in patients and mice with HD than those in controls. The up-regulation of Gal3 in HD mice occurred before motor impairment, and its level remained high in microglia throughout disease progression. The cell-autonomous up-regulated Gal3 formed puncta in damaged lysosomes and contributed to inflammation through NFκB- and NLRP3 inflammasome-dependent pathways. Knockdown of Gal3 suppressed inflammation, reduced mHTT aggregation, restored neuronal DARPP32 levels, ameliorated motor dysfunction, and increased survival in HD mice. Thus, suppression of Gal3 ameliorates microglia-mediated pathogenesis, which suggests that Gal3 is a novel druggable target for HD.


Asunto(s)
Encéfalo/patología , Galectina 3/metabolismo , Enfermedad de Huntington/patología , Microglía/patología , Adulto , Animales , Proteínas Sanguíneas , Encéfalo/citología , Encéfalo/ultraestructura , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Galectina 3/sangre , Galectina 3/genética , Galectinas , Técnicas de Silenciamiento del Gen , Humanos , Enfermedad de Huntington/sangre , Enfermedad de Huntington/diagnóstico , Inflamasomas/metabolismo , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Ratones , Microglía/citología , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Regulación hacia Arriba
10.
Mol Neurobiol ; 55(12): 8936-8952, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29616397

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and synaptic dysfunction. Adenosine is an important homeostatic modulator that controls the bioenergetic network in the brain through regulating receptor-evoked signaling pathways, bioenergetic machineries, and epigenetic-mediated gene regulation. Equilibrative nucleoside transporter 1 (ENT1) is a major adenosine transporter that recycles adenosine from the extracellular space. In the present study, we report that a small adenosine analogue (designated J4) that inhibited ENT1 prevented the decline in spatial memory in an AD mouse model (APP/PS1). Electrophysiological and biochemical analyses further demonstrated that chronic treatment with J4 normalized the impaired basal synaptic transmission and long-term potentiation (LTP) at Schaffer collateral synapses as well as the aberrant expression of synaptic proteins (e.g., NR2A and NR2B), abnormal neuronal plasticity-related signaling pathways (e.g., PKA and GSK3ß), and detrimental elevation in astrocytic A2AR expression in the hippocampus and cortex of APP/PS1 mice. In conclusion, our findings suggest that modulation of adenosine homeostasis by J4 is beneficial in a mouse model of AD. Our study provides a potential therapeutic strategy to delay the progression of AD.


Asunto(s)
Adenosina/uso terapéutico , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Tranportador Equilibrativo 1 de Nucleósido/antagonistas & inhibidores , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/fisiopatología , Plasticidad Neuronal , Presenilina-1/metabolismo , Adenosina/farmacología , Enfermedad de Alzheimer/patología , Animales , Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Placa Amiloide/patología , Placa Amiloide/fisiopatología , Receptor de Adenosina A2A/metabolismo , Transmisión Sináptica/efectos de los fármacos
11.
Sci Rep ; 6: 22529, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26932446

RESUMEN

The calcium-sensitive type VI adenylyl cyclase (AC6) is a membrane-bound adenylyl cyclase (AC) that converts ATP to cAMP under stimulation. It is a calcium-inhibited AC and integrates negative inputs from Ca(2+) and multiple other signals to regulate the intracellular cAMP level. In the present study, we demonstrate that AC6 functions upstream of CREB and negatively controls neuronal plasticity in the hippocampus. Genetic removal of AC6 leads to cyclase-independent and N-terminus of AC6 (AC6N)-dependent elevation of CREB expression, and enhances the expression of GluN2B-containing NMDA receptors in hippocampal neurons. Consequently, GluN2B-dependent calcium signaling and excitatory postsynaptic current, long-term depression, and spatial reversal learning are enhanced in the hippocampus of AC6(-/-) mice without altering the gross anatomy of the brain. Together, our results suggest that AC6 negatively regulates neuronal plasticity by modulating the levels of CREB and GluN2B in the hippocampus.


Asunto(s)
Adenilil Ciclasas/metabolismo , Aprendizaje , Depresión Sináptica a Largo Plazo/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Adenilil Ciclasas/genética , Animales , Hipocampo/enzimología , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Exp Neurol ; 248: 10-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23726959

RESUMEN

Visceral functions are regulated by a basal sympathetic nerve discharge (SND), also known as 'sympathetic tone'. We demonstrate herein that AC6 existed in tyrosine hydroxylase-positive rostral ventrolateral medulla neurons in the brainstem. Adenylyl cyclase (AC) assays showed lower basal and pituitary adenylate cyclase-activating peptide-evoked AC activities in the brainstem of AC6-null mice, indicating that AC6 is a prominent AC isozyme in the brainstem. Furthermore, two independent lines of AC6-null mice exhibited a much higher SND, recorded from splanchnic sympathetic nerves of neonatal brainstem-spinal cord preparations, than wildtype mice. An assay of urine noradrenaline confirmed this observation. Collectively, AC6 plays a critical role in the regulation of sympathetic tone.


Asunto(s)
Adenilil Ciclasas/metabolismo , Tronco Encefálico/metabolismo , Médula Espinal/metabolismo , Nervios Esplácnicos/metabolismo , Sistema Nervioso Simpático/metabolismo , Adenilil Ciclasas/genética , Animales , Animales Recién Nacidos , Ratones , Ratones Noqueados , Norepinefrina/orina
13.
Mol Cell Biol ; 31(24): 4874-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21986494

RESUMEN

3'-5'-Cyclic AMP (cAMP) is an important second messenger which regulates neurite outgrowth. We demonstrate here that type VI adenylyl cyclase (AC6), an enzyme which catalyzes cAMP synthesis, regulates neurite outgrowth by direct interaction with a binding protein (Snapin) of Snap25 at the N terminus of AC6 (AC6-N). We first showed that AC6 expression increased during postnatal brain development. In primary hippocampal neurons and Neuro2A cells, elevated AC6 expression suppressed neurite outgrowth, whereas the downregulation or genetic removal of AC6 promoted neurite extension. An AC6 variant (AC6-N5) that contains the N terminus of AC5 had no effect, indicating the importance of AC6-N. The downregulation of endogenous Snapin or the overexpression of a Snapin mutant (Snap(Δ33-51)) that does not bind to AC6, or another Snapin mutant (Snapin(S50A)) that does not interact with Snap25, reversed the inhibitory effect of AC6. Pulldown assays and immunoprecipitation-AC assays revealed that the complex formation of AC6, Snapin, and Snap25 is dependent on AC6-N and the phosphorylation of Snapin. The overexpression of Snap25 completely reversed the action of AC6. Collectively, in addition to cAMP production, AC6 plays a complex role in modulating neurite outgrowth by redistributing localization of the SNARE apparatus via its interaction with Snapin.


Asunto(s)
Adenilil Ciclasas/metabolismo , Neuritas/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Adenilil Ciclasas/genética , Animales , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Regulación hacia Abajo , Electroforesis en Gel de Poliacrilamida , Regulación del Desarrollo de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Noqueados , Mutación , Factores de Crecimiento Nervioso/análisis , Fosforilación , Plásmidos , Ratas , Ratas Sprague-Dawley , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Proteínas de Transporte Vesicular/genética
14.
J Cell Biol ; 194(2): 209-27, 2011 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-21768291

RESUMEN

Adenosine monophosphate-activated protein kinase (AMPK) is a major energy sensor that maintains cellular energy homeostasis. Huntington's disease (HD) is a neurodegenerative disorder caused by the expansion of CAG repeats in the huntingtin (Htt) gene. In this paper, we report that activation of the α1 isoform of AMPK (AMPK-α1) occurred in striatal neurons of humans and mice with HD. Overactivation of AMPK in the striatum caused brain atrophy, facilitated neuronal loss, and increased formation of Htt aggregates in a transgenic mouse model (R6/2) of HD. Such nuclear accumulation of AMPK-α1 was activity dependent. Prevention of nuclear translocation or inactivation of AMPK-α1 ameliorated cell death and down-regulation of Bcl2 caused by mutant Htt (mHtt). Conversely, enhanced expression of Bcl2 protected striatal cells from the toxicity evoked by mHtt and AMPK overactivation. These data demonstrate that aberrant activation of AMPK-α1 in the nuclei of striatal cells represents a new toxic pathway induced by mHtt.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Núcleo Celular/metabolismo , Enfermedad de Huntington/metabolismo , Neostriado/metabolismo , Neostriado/patología , Degeneración Nerviosa/metabolismo , Transporte Activo de Núcleo Celular , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Humanos , Proteína Huntingtina , Enfermedad de Huntington/enzimología , Ratones , Ratones Transgénicos , Mutación , Degeneración Nerviosa/enzimología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
15.
PLoS One ; 4(11): e7868, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19924295

RESUMEN

BACKGROUND: Normal-pressure hydrocephalus (NPH) is a neurodegenerative disorder that usually occurs late in adult life. Clinically, the cardinal features include gait disturbances, urinary incontinence, and cognitive decline. METHODOLOGY/PRINCIPAL FINDINGS: Herein we report the characterization of a novel mouse model of NPH (designated p23-ST1), created by N-ethyl-N-nitrosourea (ENU)-induced mutagenesis. The ventricular size in the brain was measured by 3-dimensional micro-magnetic resonance imaging (3D-MRI) and was found to be enlarged. Intracranial pressure was measured and was found to fall within a normal range. A histological assessment and tracer flow study revealed that the cerebral spinal fluid (CSF) pathway of p23-ST1 mice was normal without obstruction. Motor functions were assessed using a rotarod apparatus and a CatWalk gait automatic analyzer. Mutant mice showed poor rotarod performance and gait disturbances. Cognitive function was evaluated using auditory fear-conditioned responses with the mutant displaying both short- and long-term memory deficits. With an increase in urination frequency and volume, the mutant showed features of incontinence. Nissl substance staining and cell-type-specific markers were used to examine the brain pathology. These studies revealed concurrent glial activation and neuronal loss in the periventricular regions of mutant animals. In particular, chronically activated microglia were found in septal areas at a relatively young age, implying that microglial activation might contribute to the pathogenesis of NPH. These defects were transmitted in an autosomal dominant mode with reduced penetrance. Using a whole-genome scan employing 287 single-nucleotide polymorphic (SNP) markers and further refinement using six additional SNP markers and four microsatellite markers, the causative mutation was mapped to a 5.3-cM region on chromosome 4. CONCLUSIONS/SIGNIFICANCE: Our results collectively demonstrate that the p23-ST1 mouse is a novel mouse model of human NPH. Clinical observations suggest that dysfunctions and alterations in the brains of patients with NPH might occur much earlier than the appearance of clinical signs. p23-ST1 mice provide a unique opportunity to characterize molecular changes and the pathogenic mechanism of NPH.


Asunto(s)
Etilnitrosourea/farmacología , Hidrocéfalo Normotenso/metabolismo , Animales , Encéfalo/patología , Trastornos del Conocimiento/diagnóstico , Modelos Animales de Enfermedad , Femenino , Humanos , Hidrocéfalo Normotenso/diagnóstico , Presión Intracraneal , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Destreza Motora , Mutagénesis , Polimorfismo de Nucleótido Simple
16.
J Cell Physiol ; 211(2): 544-50, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17323380

RESUMEN

Progesterone is an endogenous immunomodulator and can suppress T-cell activation during pregnancy. We have previously shown that the non-genomic effects of progesterone, especially acidification, are exerted via plasma membrane sites and suppress cellular genomic responses to mitogens. This study aimed to show that acidification is due to a non-genomic inhibition of Na(+)/H(+)-exchange 1 (NHE1) by progesterone and correlate this with immunosuppressive phytohemagglutinin (PHA)-induced T-cell proliferation. The presence of amiloride-sensitive NHE 1 was identified in T cells. The activity of NHE1 was inhibited by progesterone but not by 20alpha-hydroxyprogesterone (20alpha-OHP). Furthermore, 20alpha-OHP was able to compete with progesterone and release the inhibitory effect on the NHE1. The inhibition of NHE1 activity by progesterone-BSA demonstrated non-genomic action via plasma membrane sites. Finally, co-stimulation with PHA and progesterone or amiloride, (5-(N, N-dimethyl)-amiloride, DMA), inhibited PHA-induced T-cell proliferation, but this inhibition did not occur with 20alpha-OHP and PHA co-stimulation. However, when DMA was applied 72 h after PHA stimulation, it was able to suppress PHA-induced T-cell proliferation. This is the first study to show that progesterone causes a rapid non-genomic inhibition of plasma membrane NHE1 activity in T cells within minutes which is released by 20alpha-OHP. The inhibition of NHE1 leads to immunosuppressive T-cell proliferation and suggests that progesterone might exert a major rapid non-genomic suppressive effect on NHE1 activity at the maternal-fetal interface in vivo and that 20alpha-OHP may possibly be able to quickly release the suppression when T cells circulated away from the interface.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Factores Inmunológicos/metabolismo , Activación de Linfocitos , Progesterona/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Linfocitos T/metabolismo , 20-alfa-Dihidroprogesterona/metabolismo , Adulto , Amilorida/análogos & derivados , Amilorida/farmacología , Unión Competitiva , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Concentración de Iones de Hidrógeno , Factores Inmunológicos/farmacología , Líquido Intracelular/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Mitógenos/farmacología , Fitohemaglutininas/farmacología , Progesterona/farmacología , ARN Mensajero/análisis , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/genética , Linfocitos T/efectos de los fármacos , Factores de Tiempo
17.
J Cell Biochem ; 99(1): 292-304, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16619256

RESUMEN

Progesterone is an endogenous immunomodulator, and can suppress T-cell activation during pregnancy. When analyzed under a genome time scale, the classic steroid receptor pathway does not have any effect on ion fluxes. Therefore, the aim of this study was to investigate whether the non-genomic effects on ion fluxes by progesterone could immunosuppress phytohemagglutinin (PHA)-induced human peripheral T-cell activation. The new findings indicated that, first, only progesterone stimulated both [Ca2+]i elevation and pHi decrease; in contrast, estradiol or testosterone stimulated [Ca2+]i elevation and hydrocortisone or dexamethasone stimulated pHi decrease. Secondly, the [Ca2+]i increase by progesterone was dependent on Ca2+ influx, and the acidification was blocked by Na+/H+ exchange (NHE) inhibitor, 3-methylsulphonyl-4-piperidinobenzoyl, guanidine hydrochloride (HOE-694) but not by 5-(N,N-dimethyl)-amiloride (DMA). Thirdly, progesterone blocked phorbol 12-myristate 13-acetate (PMA) or PHA-induced alkalinization, but PHA did not prevent progesterone-induced acidification. Fourthly, progesterone did not induce T-cell proliferation; however, co-stimulation progesterone with PHA was able to suppress PHA-induced IL-2 or IL-4 secretion and proliferation. When progesterone was applied 72 h after PHA stimulation, progesterone could suppress PHA-induced T-cell proliferation. Finally, immobilization of progesterone by conjugation to a large carrier molecule (BSA) also stimulated a rapid [Ca2+]i elevation, pHi decrease, and suppressed PHA-induced proliferation. These results suggested that the non-genomic effects of progesterone, especially acidification, are exerted via plasma membrane sites and suppress the genomic responses to PHA. Progesterone might act directly through membrane specific nonclassical steroid receptors to cause immunomodulation and suppression of T-cell activation during pregnancy.


Asunto(s)
Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Fitohemaglutininas/farmacología , Progesterona/farmacología , Linfocitos T/efectos de los fármacos , Adulto , Calcio/metabolismo , Calcio/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Genoma Humano , Humanos , Concentración de Iones de Hidrógeno , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Masculino , Esteroides/farmacología , Linfocitos T/fisiología , Acetato de Tetradecanoilforbol/farmacología , Timidina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA