Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vet Intern Med ; 37(6): 2230-2240, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37728198

RESUMEN

BACKGROUND: It is unknown if enrofloxacin accumulates in plasma of cats with reduced kidney function. HYPOTHESIS: To determine if enrofloxacin and its active metabolite ciprofloxacin have reduced clearance in azotemic cats. ANIMALS: Thirty-four cats hospitalized for clinical illness with variable degree of kidney function. METHODS: Prospective study. After enrofloxacin (dose 5 mg/kg) administration to cats, sparse blood sampling was used to obtain 2 compartment population pharmacokinetic results using nonlinear mixed-effects modeling. Plasma enrofloxacin and ciprofloxacin concentrations were measured and summed to obtain the total fluoroquinolone concentration. A model of ciprofloxacin metabolism from enrofloxacin was created and evaluated for covariate effects on clearance, volume of distribution, and the metabolic rate of ciprofloxacin generation from enrofloxacin. RESULTS: Body weight was the only covariate found to affect total fluoroquinolone volume of distribution (effect 1.63, SE 0.19, P < .01) and clearance (effect 1.63, SE 0.27, P < .01). Kidney function did not have a significant effect on total fluoroquinolone clearance (median 440.8 mL/kg/h (range 191.4-538.0 mL/kg/h) in cats with normal kidney function, 365.8 mL/kg/h (range 89.49-1092.0 mL/kg/h) in cats with moderate kidney dysfunction, and 308.5 mL/kg/h (range 140.20-480.0 mL/kg/h) in cats with severe kidney dysfunction (P = .64). Blood urea nitrogen concentration influenced the metabolic generation of ciprofloxacin from enrofloxacin (effect 0.51, SE 0.08, P < .01), but other markers of kidney function did not. CONCLUSIONS AND CLINICAL IMPORTANCE: Adjustment of enrofloxacin dosage is not indicated for azotemic cats.


Asunto(s)
Ciprofloxacina , Fluoroquinolonas , Gatos , Animales , Enrofloxacina , Inyecciones Intravenosas/veterinaria , Estudios Prospectivos , Riñón
2.
J Alzheimers Dis ; 93(2): 605-619, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37066917

RESUMEN

BACKGROUND: Deprivation of extracellular serotonin has been linked to cognitive decline and neuropsychiatric disturbances in Alzheimer's disease (AD). However, despite degeneration of serotonin-producing neurons, whether serotonin release is affected in AD-sensitive brain regions is unknown. OBJECTIVE: This study investigated the impact of mitochondrial dysfunction in decreased hippocampal serotonin release in AD amyloidosis mouse model 5xFAD mice. METHODS: Electrochemical assays were applied to examine hippocampal serotonin release. We also employed multidisciplinary techniques to determine the role of oligomeric amyloid-ß (Aß) in hippocampal mitochondrial deficits and serotonin release deficiency. RESULTS: 5xFAD mice exhibited serotonin release decrease and relatively moderate downregulation of serotonergic fiber density as well as serotonin content in the hippocampal region. Further experiments showed an inhibitory effect of oligomeric amyloid-ß (Aß) on hippocampal serotonin release without affecting the density of serotonergic fibers. Pharmaceutical uncoupling of mitochondrial oxidative phosphorylation (OXPHOS) disrupted hippocampal serotonin release in an ex vivo setting. This echoes the mitochondrial defects in serotonergic fibers in 5xFAD mice and oligomeric Aß-challenged primary serotonergic neuron cultures and implicates a link between mitochondrial dysfunction and serotonin transmission defects in AD-relevant pathological settings. CONCLUSION: The most parsimonious interpretation of our findings is that mitochondrial dysfunction is a phenotypic change of serotonergic neurons, which potentially plays a role in the development of serotonergic failure in AD-related conditions.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/patología , Serotonina/metabolismo , Ratones Transgénicos , Péptidos beta-Amiloides/metabolismo , Hipocampo/patología , Mitocondrias/metabolismo , Modelos Animales de Enfermedad
3.
ACS Chem Neurosci ; 13(15): 2315-2337, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35833925

RESUMEN

Huntington's disease is a progressive and lethal neurodegenerative disease caused by an increased CAG repeat mutation in exon 1 of the huntingtin gene (mutant huntingtin). Current drug treatments provide only limited symptomatic relief without impacting disease progression. Previous studies in our lab and others identified the abnormal binding of mutant huntingtin protein with calmodulin, a key regulator of calcium signaling. Disrupting the abnormal binding of mutant huntingtin to calmodulin reduces perturbations caused by mutant huntingtin in cell and mouse models of Huntington's disease and importantly normalizes receptor-stimulated calcium release. Using a series of high-throughput in vitro and cell-based screening assays, we identified numerous small-molecule hits that disrupt the binding of mutant huntingtin to calmodulin and demonstrate protective effects. Iterative optimization of one hit resulted in nontoxic, selective compounds that are protective against mutant huntingtin cytotoxicity and normalized receptor-stimulated intracellular calcium release in PC12 cell models of Huntington's disease. Importantly, the compounds do not work by reducing the levels of mutant huntingtin, allowing this strategy to complement future molecular approaches to reduce mutant huntingtin expression. Our novel scaffold will serve as a prototype for further drug development in Huntington's disease. These studies indicate that the development of small-molecule compounds that disrupt the binding of mutant huntingtin to calmodulin is a promising approach for the advancement of therapeutics to treat Huntington's disease.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Animales , Calcio/metabolismo , Calmodulina/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo
4.
Int J Mol Sci ; 22(24)2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34947973

RESUMEN

Serotonin 1A receptors (5-HT1ARs) are implicated in the control of mood, cognition, and memory and in various neuropsychiatric disorders such as depression and anxiety. As such, understanding the regulation of 5-HT1ARs will inform the development of better treatment approaches. We previously demonstrated 5-HT1ARs are SUMOylated by SUMO1 in the rat brain. Agonist stimulation increased SUMOylation and was further enhanced when combined with 17ß-estradiol-3-benzoate (EB), which are treatments that cause the transient and prolonged desensitization of 5-HT1AR signaling, respectively. In the current study, we identified the protein inhibitor of activated STAT (PIAS)xα as the enzyme that facilitates SUMOylation, and SENP2 as the protein that catalyzes the deSUMOylation of 5-HT1ARs. We demonstrated that PIASxα significantly increased in the membrane fraction of rats co-treated with EB and an agonist, compared to either the EB-treated or vehicle-treated groups. The acute treatment with an agonist alone shifted the location of SENP2 from the membrane to the cytoplasmic fraction, but it has little effect on PIASxα. Hence, two separate mechanisms regulate SUMOylation and the activity of 5-HT1ARs by an agonist and EB. The effects of EB on 5-HT1AR SUMOylation and signaling may be related to the higher incidence of mood disorders in women during times with large fluctuations in estrogens. Targeting the SUMOylation of 5-HT1ARs could have important clinical relevance for the therapy for several neuropsychiatric disorders in which 5-HT1ARs are implicated.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Estradiol/análogos & derivados , Proteínas Inhibidoras de STAT Activados/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Estradiol/administración & dosificación , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratas , Sumoilación/efectos de los fármacos , Regulación hacia Arriba
5.
Med Chem Res ; 29(7): 1187-1198, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33642842

RESUMEN

The aberrant protein-protein interaction between calmodulin and mutant huntingtin protein in Huntington's disease patients has been found to contribute to Huntington's disease progression. A high-throughput screen for small molecules capable of disrupting this interaction revealed a sultam series as potent small-molecule disruptors. Diversification of the sultam scaffold afforded a set of 24 analogs or further evaluation. Several structure-activity trends within the analog set were found, most notably a negligible effect of absolute stereochemistry and a strong beneficial correlation with electron-withdrawing aromatic substituents. The most promising analogs were profiled for off-target effects at relevant kinases and, ultimately, one candidate molecule was evaluated for neuroprotection in a neuronal cell model of Huntington's disease.

6.
Biomolecules ; 9(11)2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31752360

RESUMEN

Finasteride (FIN) is the prototypical inhibitor of steroid 5α-reductase (5αR), the enzyme that catalyzes the rate-limiting step of the conversion of progesterone and testosterone into their main neuroactive metabolites. FIN is clinically approved for the treatment of benign prostatic hyperplasia and male baldness; while often well-tolerated, FIN has also been shown to cause or exacerbate psychological problems in vulnerable subjects. Evidence on the psychological effects of FIN, however, remains controversial, in view of inconsistent clinical reports. Here, we tested the effects of FIN in a battery of tests aimed at capturing complementary aspects of mood regulation and stress reactivity in rats. FIN reduced exploratory, incentive, prosocial, and risk-taking behavior; furthermore, it decreased stress coping, as revealed by increased immobility in the forced-swim test (FST). This last effect was also observed in female and orchiectomized male rats, suggesting that the mechanism of action of FIN does not primarily reflect changes in gonadal steroids. The effects of FIN on FST responses were associated with a dramatic decrease in corticotropin release hormone (CRH) mRNA and adrenocorticotropic hormone (ACTH) levels. These results suggest that FIN impairs stress reactivity and reduces behavioral activation and impulsive behavior by altering the function of the hypothalamus-pituitary-adrenal (HPA) axis.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Hormona Adrenocorticotrópica/metabolismo , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Finasterida/farmacología , Estrés Psicológico , Afecto/efectos de los fármacos , Animales , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/patología , Ratas , Ratas Long-Evans , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/patología
7.
Med One ; 3(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30701197

RESUMEN

Transglutaminases (TGs) and especially TG2 play important roles in neurotransmitter and receptor signaling pathways. Three different mechanisms by which TG2 interacts with neurotransmitter and receptor signaling systems will be discussed in this review. The first way in which TG2 interacts with receptor signaling is via its function as a guanine nucleotide binding protein (G-protein) coupling to G-protein coupled receptors (GPCRs) to activate down-stream signaling pathways. TG2 can exist in a least two conformations, a closed GTP-bound conformation and an open calcium-bound conformation. In the closed GTP-bound conformation, TG2 is capable of functioning as a G-protein for GPCRs. In the open calcium-bound conformation, TG2 catalyzes a transamidation reaction cross-linking proteins or catalyzing the covalent binding of a mono- or polyamine to a protein. The second mechanism is regulation of the transamidation reaction catalyzed by TG2 via receptor stimulation which can increase local calcium concentrations and thereby increase transamidation reactions. The third way in which TG2 plays a role in neurotransmitter and receptor signaling systems is via its use of monoamine neurotransmitters as a substrate. Monoamine neurotransmitters including serotonin can be substrates for transamidation to a protein often a small G-protein (also known as a small GTPase) resulting in activation of the small G-protein. The transamidation of a monoamine neurotransmitter or serotonin has been designated as monoaminylation or more specifically serotonylation, respectively. Other proteins are also targets for monoaminylation such as fibronectin and cytoskeletal proteins. These receptor and neurotransmitter-regulated reactions by TG2 play roles in physiological and key pathophysiological processes.

8.
Neuropharmacology ; 117: 93-105, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28161375

RESUMEN

Regulation of dendritic spines is an important component of synaptic function and plasticity whereas dendritic spine dysregulation is related to several psychiatric and neurological diseases. In the present study, we tested the hypothesis that serotonin (5-HT)2A/2C receptor-induced Rho family transamidation and activation regulates dendritic spine morphology and that activation of multiple types of receptors can induce transglutaminase (TGase)-catalyzed transamidation of small G proteins. We previously reported a novel 5-HT2A receptor downstream effector, TGase-catalyzed serotonylation of the small G protein Rac1 in A1A1v cells, a rat embryonic cortical cell line. We now extend these findings to rat primary cortical cultures which develop dendritic spines; stimulation of 5-HT2A/2C receptors increased transamidation of Rac1 and Cdc42, but not RhoA. Inhibition of TGases significantly decreased transamidation and activation of Rac1 and Cdc42, suggesting that transamidation led to their activation. In primary cortical cultures, stimulation of 5-HT2A/2C receptors by 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) caused a transient dendritic spine enlargement, which was blocked by TGase inhibition. Stimulation of both 5-HT2A and 5-HT2C receptors contributed to DOI-induced Rac1 transamidation in primary cortical cultures as demonstrated by selective antagonists. Furthermore, stimulation of muscarinic acetylcholine receptors and NMDA receptors also increased TGase-catalyzed Rac1 activation in SH-SY5Y cells and N2a cells, respectively. Receptor-stimulated TGase-catalyzed transamidation of Rac1 occurs at Q61, a site previously reported to be important in the inactivation of Rac1. These studies demonstrate that TGase-catalyzed transamidation and activation of small G proteins results from stimulation of multiple types of receptors and this novel signaling pathway can regulate dendritic spine morphology and plasticity.


Asunto(s)
Espinas Dendríticas/fisiología , Receptor de Serotonina 5-HT2A/fisiología , Transglutaminasas/fisiología , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Anfetaminas/farmacología , Animales , Humanos , Ratones , Cultivo Primario de Células , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Transglutaminasas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
9.
Behav Brain Res ; 294: 7-16, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26210937

RESUMEN

Behavioral testing of mouse models of Huntington's disease (HD) is a key component of preclinical assessment for potential pharmacological intervention. An open field with a force plate floor was used to quantify numerous spontaneous behaviors in a slowly progressing model of HD. CAG140 (+/+, +/-, -/-) male and female mice were compared in a longitudinal study from 6 to 65 weeks of age. Distance traveled, wall rears, wall rear duration, number of low mobility bouts, in-place movements, number of high velocity runs, and gait parameters (stride rate, stride length, and velocity) were extracted from the ground reaction forces recorded in 20-min actometer sessions. Beginning at 11 weeks, HD mice (both +/- and +/+) were consistently hypoactive throughout testing. Robust hypoactivity at 39 weeks of age was not accompanied by gait disturbances. By 52 and 65 weeks of age the duration of wall rears increased and in-place tremor-like movements emerged at 65 weeks of age in the +/+, but not in the +/- HD mice. Taken together, these results suggest that hypoactivity preceding frank motor dysfunction is a characteristic of CAG140 mice that may correspond to low motivation to move seen clinically in the premanifest/prediagnostic stage in human HD. The results also show that the force plate method provides a means for tracking the progression of behavioral dysfunction in HD mice beyond the stage when locomotion is lost while enabling quantification of tremor-like and similar in-place behaviors without a change in instrumentation. Use of force plate actometry also minimizes testing-induced enrichment effects when batteries of different tests are carried out longitudinally.


Asunto(s)
Acelerometría/instrumentación , Acelerometría/métodos , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Enfermedad de Huntington/fisiopatología , Animales , Fenómenos Biomecánicos , Peso Corporal , Progresión de la Enfermedad , Femenino , Marcha/fisiología , Técnicas de Sustitución del Gen , Estudios Longitudinales , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Temblor/fisiopatología
10.
ACS Chem Neurosci ; 6(7): 961-9, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25615632

RESUMEN

Although serotonin was discovered over 65 years ago, it has been only within the past decade that serotonin was found to be involved in a covalent post-translational modification to proteins. The enzyme transglutaminase catalyzes the transamidation of serotonin to a protein-bound glutamine residue; the amino group of serotonin is covalently bound to the gamma carboxamide of glutamine. The term serotonylation is used to describe this transamidation reaction to serotonin. Not only can serotonin be a substrate for transamidation to proteins but also other monoamine neurotransmitters are substrates including histamine, dopamine, and noradrenaline. The term monoaminylation has been coined to describe the transamidation of monoamines to protein substrates. Small G proteins have emerged as the most common substrate for monoaminylation and are activated by this post-translational modification. Fibronectin and cytoskeletal proteins are also substrates for monoaminylation. Serotonylation and monoaminylation are involved in a number of physiological functions, including platelet activation, insulin release, smooth muscle contraction, and regulation of membrane localization of the serotonin transporter. Stimulation of 5-HT2A receptors increases serotonylation and activates the small G protein Rac1, which plays a role in dendritic spine regulation. Monoaminylation is implicated in pathophysiological processes as well such as diabetes and hypertension. The availability of monoamines for monoaminylation is altered by antidepressants that target serotonin transporters, noradrenaline transporters, or the enzymatic degradation of monoamines as well as drugs of abuse such as cocaine and amphetamines. Further research on monoaminylation is needed to elucidate its physiological and pathophysiological roles and to explore monoaminylation as a novel target for drug therapy.


Asunto(s)
Serotonina/metabolismo , Transglutaminasas/metabolismo , Animales , Humanos , Neuronas/metabolismo
11.
Neuroendocrinology ; 100(2-3): 228-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25402859

RESUMEN

Hyperactivity of the hypothalamic-pituitary-adrenal axis is a consistent biological characteristic of depression, and response normalization coincides with clinical responsiveness to antidepressant medications. Desensitization of serotonin 1A receptor (5-HT1AR) signaling in the hypothalamic paraventricular nucleus of the hypothalamus (PVN) follows selective serotonin reuptake inhibitor (SSRI) antidepressant treatment and contributes to the antidepressant response. Estradiol alone produces a partial desensitization of 5-HT1AR signaling and synergizes with SSRIs to result in a complete and more rapid desensitization than with SSRIs alone as measured by a decrease in the oxytocin and adrenocorticotrophic hormone (ACTH) responses to 5-HT1AR stimulation. G protein-coupled estrogen receptor 1 (GPER1) is necessary for estradiol-induced desensitization of 5-HT1AR signaling, although the underlying mechanisms are still unclear. We now find that stimulation of GPER1 with the selective agonist G-1 and nonselective stimulation of estrogen receptors dramatically alter isoform expression of a key component of the 5-HT1AR signaling pathway, RGSz1, a GTPase-activating protein selective for Gαz, the Gα subunit necessary for 5-HT1AR-mediated hormone release. RGSz1 isoforms are differentially glycosylated, SUMOylated, and phosphorylated, and differentially distributed in subcellular organelles. High-molecular-weight RGSz1 is SUMOylated and glycosylated, localized to the detergent-resistant microdomain (DRM) of the cell membrane, and increased by estradiol and G-1 treatment. Because activated Gαz also localizes to the DRM, increased DRM-localized RGSz1 by estradiol and G-1 could reduce Gαz activity, functionally uncoupling 5-HT1AR signaling. Peripheral G-1 treatment produced a partial reduction in oxytocin and ACTH responses to 5-HT1AR stimulation similar to direct injections into the PVN. Together, these results identify GPER1 and RGSz1 as novel targets for the treatment of depression.


Asunto(s)
Hipotálamo/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas RGS/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Membrana Celular/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Ciclopentanos/farmacología , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Glicosilación , Hipotálamo/efectos de los fármacos , Orgánulos/metabolismo , Fosforilación , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Quinolinas/farmacología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal , Sumoilación
12.
Psychoneuroendocrinology ; 38(11): 2542-53, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23786880

RESUMEN

Sumoylation is a recently described post-translational modification and only a few sumoylated neurotransmitter receptors are known. Through the present studies, we discovered that serotonin1A receptors (5-HT1A-Rs) can be sumoylated by SUMO1 (small-ubiquitin-related modifier 1) protein. The SUMO1-5-HT1A-R is ∼55kDa, is located in the membrane fraction, but not the cytosol, and is distributed in all of the brain regions expressing 5-HT1A-Rs examined. Acute stimulation of 5-HT1A-Rs significantly increased SUMO1-5-HT1A-R in rat hypothalamus. Pre-treatment with estradiol for 2 days, which causes a partial desensitization of 5-HT1A-R signaling, potentiated agonist-induced increases in SUMO1-5-HT1A-Rs in the hypothalamus of ovariectomized rats. Using discontinuous gradient centrifugation followed by digitonin treatment, we found that the majority of SUMO1-5-HT1A-Rs is co-localized with endoplasmic-reticulum and trans-Golgi-network markers. Although a small proportion of SUMO1-5-HT1A-Rs are located in the detergent resistant microdomain (DRM) that contain active G-protein coupled receptors, their distribution was different from that of the Gαz protein that couples to the receptors. These data suggest that the SUMO1-5-HT1A-Rs are an inactive form of 5-HT1A-Rs, a finding further supported by results showing minimal 5-HT1A-R agonist binding to SUMO1-5-HT1A-Rs. Furthermore, SUMO1-5-HT1A-Rs in the DRM were increased by treatment with a 5-HT1A-R agonist, 8-OH-DPAT ((+)8-hydroxy-2-dipropylaminotetralin). Together, these data suggest that sumoylation of 5-HT1A-Rs may be related to 5-HT1A-R trafficking and internalization, which may contribute to 5-HT1A-R desensitization. Since 5-HT1A-Rs play an important role in mood regulation, the present results significantly impact on the understanding of the pathogenesis of affective disorders and development of better therapeutic approaches for these diseases.


Asunto(s)
8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Estradiol/farmacología , Receptor de Serotonina 5-HT1A/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Fracciones Subcelulares/metabolismo , Sumoilación/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Sinergismo Farmacológico , Retículo Endoplásmico/metabolismo , Estrógenos/farmacología , Femenino , Fluoxetina/farmacología , Aparato de Golgi/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
13.
Psychoneuroendocrinology ; 38(7): 1145-57, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23219224

RESUMEN

A major problem with current anti-depressant therapy is that it takes on average 6-7 weeks for remission. Since desensitization of serotonin (5-HT)1A receptor signaling contributes to the anti-depressive response, acceleration of the desensitization may reduce this delay in response to antidepressants. The purpose of the present study was to test the hypothesis that estradiol accelerates fluoxetine-induced desensitization of 5-HT1A receptor signaling in the paraventricular nucleus of the hypothalamus (PVN) of rats, via alterations in components of the 5-HT1A receptor signaling pathway. Ovariectomized rats were injected with estradiol and/or fluoxetine, then adrenocorticotropic hormone (ACTH) and oxytocin responses to a 5-HT1A receptor agonist (+)-8-hydroxy-2-dipropylaminotetralin (8-OH-DPAT) were examined to assess the function of 5-HT1A receptors in the PVN. Treatment with estradiol for either 2 or 7 days or fluoxetine for 2 days produced at most a partial desensitization of 5-HT1A receptor signaling, whereas 7 days of fluoxetine produced full desensitization. Combined treatment with estradiol and fluoxetine for 2 days produced nearly a full desensitization, demonstrating an accelerated response compared to either treatment alone. With two days of combined treatments, estradiol prevented the fluoxetine-induced increase in 5-HT1A receptor protein, which could contribute to the more rapid desensitization. Furthermore, EB treatment for 2 days decreased the abundance of the 35 kD Gαz protein which could contribute to the desensitization response. We found two isoforms of Gαz proteins with molecular mass of 35 and 33 kD, which differentially distributed in the detergent resistant microdomain (DRM) and in Triton X-100 soluble membrane region, respectively. The 35 kD Gαz proteins in the DRM can be sumoylated by SUMO1. Stimulation of 5-HT1A receptors with 8-OH-DPAT increases the sumoylation of Gαz proteins and reduces the 33 kD Gαz proteins, suggesting that these responses may be related to the desensitization of 5-HT1A receptors. Treatment with estradiol for 2 days also reduced the levels of the G-protein coupled estrogen receptor GPR30, possibly limiting to the ability of estradiol to produce only a partial desensitization response. These data provide evidence that estradiol may be effective as a short-term adjuvant to SSRIs to accelerate the onset of therapeutic effects.


Asunto(s)
Estradiol/farmacología , Fluoxetina/farmacología , Receptor de Serotonina 5-HT1A/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Hormona Adrenocorticotrópica/sangre , Animales , Interacciones Farmacológicas , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Oxitocina/sangre , Ratas , Ratas Sprague-Dawley , Agonistas de Receptores de Serotonina
15.
Psychopharmacology (Berl) ; 213(2-3): 403-12, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20717650

RESUMEN

RATIONALE: Serotonin and especially serotonin 2A (5-HT(2A)) receptor signaling are important in the etiology and treatment of schizophrenia and affective disorders. We previously reported a novel 5-HT(2A) receptor effector, increased transglutaminase (TGase)-catalyzed transamidation, and activation of the small G protein Rac1 in A1A1v cells, a rat embryonic cortical cell line. OBJECTIVES: In this study, we explore the signaling pathway involved in 5-HT(2A) receptor-mediated Rac1 transamidation. METHODS: A1A1v cells were pretreated with pharmacological inhibitors of phospholipase C (PLC) or calmodulin (CaM), and then stimulated by the 5-HT(2A) receptor agonist, 2,5-dimethoxy-4-iodoamphetamine (DOI). Intracellular Ca(2+) concentration and TGase-modified Rac1 transamidation were monitored. The effect of manipulation of intracellular Ca(2+) by a Ca(2+) ionophore or a chelating agent on Rac1 transamidation was also evaluated. RESULTS: In cells pretreated with a PLC inhibitor U73122, DOI-stimulated increases in the intracellular Ca(2+) concentration and TGase-modified Rac1 were significantly attenuated as compared to those pretreated with U73343, an inactive analog. The membrane-permeant Ca(2+) chelator, BAPTA-AM strongly reduced TGase-catalyzed Rac1 transamidation upon DOI stimulation. Conversely, the Ca(2+) ionophore ionomycin, at a concentration that induced an elevation of cytosolic Ca(2+) to a level comparable to cells treated with DOI, produced an increase in TGase-modified Rac1 without 5-HT(2A) receptor activation. Moreover, the CaM inhibitor W-7, significantly decreased Rac1 transamidation in a dose-dependent manner in DOI-treated cells. CONCLUSIONS: These results indicate that 5-HT(2A) receptor-coupled PLC activation and subsequent Ca(2+) and CaM signaling are necessary for TGase-catalyzed Rac1 transamidation, and an increase in intracellular Ca(2+) is sufficient to induce Rac1 transamidation.


Asunto(s)
Calcio/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Transglutaminasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Anfetaminas/farmacología , Animales , Calmodulina/metabolismo , Línea Celular , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Estrenos/farmacología , Ionomicina/farmacología , Pirrolidinonas/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Fosfolipasas de Tipo C/metabolismo , Proteína de Unión al GTP rac1/efectos de los fármacos
16.
Psychoneuroendocrinology ; 35(7): 1023-33, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20138435

RESUMEN

Estradiol regulates serotonin 1A (5-HT(1A)) receptor signaling. Since desensitization of 5-HT(1A) receptors may be an underlying mechanism by which selective serotonin reuptake inhibitors (SSRIs) mediate their therapeutic effects and combining estradiol with SSRIs enhances the efficacy of the SSRIs, it is important to determine which estrogen receptors are capable of desensitizating 5-HT(1A) receptor function. We previously demonstrated that selective activation of the estrogen receptor, GPR30, desensitizes 5-HT(1A) receptor signaling in rat hypothalamic paraventricular nucleus (PVN). However, since estrogen receptor-beta (ERbeta), is highly expressed in the PVN, we investigated the role of ERbeta in estradiol-induced desensitization of 5-HT(1A) receptor signaling. We first showed that a selective ERbeta agonist, diarylpropionitrile (DPN) has a 100-fold lower binding affinity than estradiol for GPR30. Administration of DPN did not desensitize 5-HT(1A) receptor signaling in rat PVN as demonstrated by agonist-stimulated hormone release. Second, we used a recombinant adenovirus containing ERbeta siRNAs to decrease ERbeta expression in the PVN. Reductions in ERbeta did not alter the estradiol-induced desensitization of 5-HT(1A) receptor signaling in oxytocin cells. In contrast, in animals with reduced ERbeta, estradiol administration, instead of producing desensitization, augmented the ACTH response to a 5-HT(1A) agonist. Combined with the results from the DPN treatment experiments, desensitization of 5-HT(1A) receptor signaling does not appear to be mediated by ERbeta in oxytocin cells, but that ERbeta, together with GPR30, may play a complex role in central regulation of 5-HT(1A)-mediated ACTH release. Determining the mechanisms by which estrogens induce desensitization may aid in the development of better treatments for mood disorders.


Asunto(s)
Estradiol/metabolismo , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/metabolismo , Animales , Estradiol/farmacología , Femenino , Nitrilos/farmacología , Oxitocina/sangre , Oxitocina/metabolismo , Propionatos/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
17.
Brain Pathol ; 20(1): 176-89, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19338577

RESUMEN

Huntington's disease (HD) is a neurodegenerative disease caused by mutant huntingtin protein containing an expanded polyglutamine tract, which may cause abnormal protein-protein interactions such as increased association with calmodulin (CaM). We previously demonstrated in HEK293 cells that a peptide containing amino acids 76-121 of CaM (CaM-peptide) interrupted the interaction between CaM and mutant huntingtin, reduced mutant huntingtin-induced cytotoxicity and reduced transglutaminase (TG)-modified mutant huntingtin. We now report that adeno-associated virus (AAV)-mediated expression of CaM-peptide in differentiated neuroblastoma SH-SY5Y cells, stably expressing an N-terminal fragment of huntingtin containing 148 glutamine repeats, significantly decreases the amount of TG-modified huntingtin and attenuates cytotoxicity. Importantly, the effect of the CaM-peptide shows selectivity, such that total TG activity is not significantly altered by expression of CaM-peptide nor is the activity of another CaM-dependent enzyme, CaM kinase II. In vitro, recombinant exon 1 of huntingtin with 44 glutamines (htt-exon1-44Q) binds to CaM-agarose; the addition of 10 microM of CaM-peptide significantly decreases the interaction of htt-exon1-44Q and CaM but not the binding between CaM and calcineurin, another CaM-binding protein. These data support the hypothesis that CaM regulates TG-catalyzed modifications of mutant huntingtin and that specific and selective disruption of the CaM-huntingtin interaction is potentially a new target for therapeutic intervention in HD.


Asunto(s)
Calcio/metabolismo , Calmodulina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Fármacos Neuroprotectores , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/farmacología , Adenoviridae/genética , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Exones , Citometría de Flujo , Vectores Genéticos , Humanos , Proteína Huntingtina , Inmunoprecipitación , Microscopía Fluorescente , Mutación/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Ácido Poliglutámico/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes , Transglutaminasas/metabolismo
18.
J Neurosci ; 29(37): 11550-9, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19759302

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder, caused by a polyglutamine expansion in the huntingtin protein (htt). Increasing evidence suggests that transglutaminase (TGase) plays a critical role in the pathophysiology of HD possibly by stabilizing monomeric, polymeric and aggregated htt. We previously reported that in HEK293 and SH-SY5Y cells expression of a calmodulin (CaM)-fragment, consisting of amino acids 76-121 of CaM, decreased binding of CaM to mutant htt, TGase-modified htt and cytotoxicity associated with mutant htt and normalized intracellular calcium release. In this study, an adeno-associated virus (AAV) that expresses the CaM-fragment was injected into the striatum of HD transgenic R6/2 mice. The CaM-fragment significantly reduced body weight loss and improved motor function as indicated by improved rotarod performance, longer stride length, lower stride frequency, fewer low mobility bouts and longer travel distance than HD controls. A small but insignificant increase in survival was observed in R6/2 mice with CaM-fragment expression. Immunoprecipitation studies show that expression of the CaM-fragment reduced TGase-modified htt in the striatum of R6/2 mice. The percentage of htt-positive nuclei and the size of intranuclear htt aggregates were reduced by the CaM-fragment without striatal volume changes. The effects of CaM-fragment appear to be selective, as activity of another CaM-dependent enzyme, CaM-dependent kinase II, was not altered. Moreover, inhibition of TGase-modified htt was substrate-specific since overall TGase activity in the striatum was not altered by treatment with the CaM-fragment. Together, these results suggest that disrupting CaM-htt interaction may provide a new therapeutic strategy for HD.


Asunto(s)
Peso Corporal/fisiología , Calmodulina/metabolismo , Cuerpo Estriado/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Análisis de Varianza , Animales , Conducta Animal/fisiología , Peso Corporal/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Proteínas de Unión al GTP/metabolismo , Marcha/genética , Regulación Enzimológica de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Enfermedad de Huntington/genética , Inmunoprecipitación/métodos , Locomoción/genética , Locomoción/fisiología , Masculino , Ratones , Ratones Transgénicos , Actividad Motora , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Prueba de Desempeño de Rotación con Aceleración Constante , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transglutaminasas/metabolismo , Expansión de Repetición de Trinucleótido/genética
19.
Int J Neuropsychopharmacol ; 12(5): 651-65, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18976543

RESUMEN

We have previously demonstrated that olanzapine-induced desensitization of 5-HT2A receptor-stimulated phospholipase C (PLC) activity is associated with increases in RGS7 protein levels both in vivo and in cells in culture, and the increase in RGS7 is dependent on activation of the JAK-STAT pathway in cells in culture. In the present study, we found that desensitization of 5-HT2A receptor-stimulated PLC activity induced by olanzapine is dependent on activation of the JAK-STAT pathway. Similar to olanzapine, clozapine-induced desensitization of 5-HT2A receptor signalling is accompanied by increases in RGS7 and activation of JAK2. Treatment with the selective 5-HT2A receptor antagonist MDL 100907 also increased RGS7 protein levels and JAK2 activation. Using a JAK2 inhibitor AG490, we found that clozapine and MDL 100907-induced increases in RGS7 are dependent on activation of the JAK-STAT pathway. Olanzapine, clozapine, and MDL 100907 treatment increased mRNA levels of RGS7. Using a chromatin immunoprecipitation assay we found STAT3 binding to the putative RGS7 promoter region. Taken together, olanzapine-induced activation of the JAK-STAT pathway, and STAT3 binding to the RGS7 gene could underlie the increase in RGS7 mRNA which could subsequently increase protein expression. Furthermore, the increase in RGS7 protein could play a role in the desensitization of 5-HT2A receptor signalling by terminating the activated Galphaq/11 proteins more rapidly. Overall, our data suggest that the complete desensitization of 5-HT2A receptor-stimulated PLC activity by olanzapine, clozapine and MDL 100907 requires activation of the JAK-STAT pathway, which in turn increases RGS7 expression probably by direct transcriptional activity of STAT3.


Asunto(s)
Benzodiazepinas/farmacología , Clozapina/farmacología , Fluorobencenos/farmacología , Quinasas Janus/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Piperidinas/farmacología , Factor de Transcripción STAT3/fisiología , Antagonistas del Receptor de Serotonina 5-HT2 , Fosfolipasas de Tipo C/fisiología , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Olanzapina , Receptor de Serotonina 5-HT2A/metabolismo
20.
Future Neurol ; 4(4): 449-467, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20161049

RESUMEN

The following review examines the role of calcium in promoting the in vitro and in vivo activation of transglutaminases in neurodegenerative disorders. Diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease exhibit increased transglutaminase activity and rises in intracellular calcium concentrations, which may be related. The aberrant activation of transglutaminase by calcium is thought to give rise to a variety of pathological moieties in these diseases, and the inhibition has been shown to have therapeutic benefit in animal and cellular models of neurodegeneration. Given the potential clinical relevance of transglutaminase inhibitors, we have also reviewed the recent development of such compounds.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...