Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell Mol Neurobiol ; 42(1): 255-263, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32865675

RESUMEN

We have previously shown that angiotensin-converting enzyme 2 (ACE2), an enzyme counterbalancing the deleterious effects of angiotensin type 1 receptor activation by production of vasodilatory peptides Angiotensin (Ang)-(1-9) and Ang-(1-7), is internalized and degraded in lysosomes following chronic Ang-II treatment. However, the molecular mechanisms involved in this effect remain unknown. In an attempt to identify the accessory proteins involved in this effect, we conducted a proteomic analysis in ACE2-transfected HEK293T cells. A single protein, fascin-1, was found to differentially interact with ACE2 after Ang-II treatment for 4 h. The interactions between fascin-1 and ACE2 were confirmed by confocal microscopy and co-immunoprecipitation. Overexpression of fascin-1 attenuates the effects of Ang-II on ACE2 activity. In contrast, downregulation of fascin-1 severely decreased ACE2 enzymatic activity. Interestingly, in brain homogenates from hypertensive mice, we observed a significant reduction of fascin-1, suggesting that the levels of this protein may change in cardiovascular diseases. In conclusion, we identified fascin-1 as an ACE2-accessory protein, interacting with the enzyme in an Ang-II dependent manner and contributing to the regulation of enzyme activity.


Asunto(s)
Actinas , Enzima Convertidora de Angiotensina 2 , Proteínas Portadoras , Proteínas de Microfilamentos , Actinas/metabolismo , Angiotensina I/metabolismo , Angiotensina II/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células HEK293 , Humanos , Ratones , Proteínas de Microfilamentos/metabolismo , Fragmentos de Péptidos/metabolismo , Proteómica
2.
Clin Sci (Lond) ; 132(14): 1513-1527, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29903768

RESUMEN

TRV027 is a biased agonist for the Angiotensin (Ang)-II type 1 receptor (AT1R), able to recruit ß-arrestin 2 independently of G-proteins activation. ß-arrestin activation in the central nervous system (CNS) was suggested to oppose the effects of Ang-II. The present study evaluates the effect of central infusion of TRV027 on arterial pressure (AP), autonomic function, baroreflex sensitivity (BRS), and peripheral vascular reactivity. Spontaneously hypertensive (SH) and Wistar Kyoto (WKY) rats were treated with TRV027 for 14 days (20 ng/h) delivered to the lateral ventricle via osmotic minipumps. Mechanistic studies were performed in HEK293T cells co-transfected with AT1R and Ang converting enzyme type 2 (ACE2) treated with TRV027 (100 nM) or Ang-II (100 nM). TRV027 infusion in SH rats (SHR) reduced AP (~20 mmHg, P<0.05), sympathetic vasomotor activity (ΔMAP = -47.2 ± 2.8 compared with -64 ± 5.1 mmHg, P<0.05) and low-frequency (LF) oscillations of AP (1.7 ± 0.2 compared with 5.8 ± 0.4 mmHg, P<0.05) compared with the SHR control group. TRV027 also increased vagal tone, improved BRS, reduced the reactivity of mesenteric arteries to Ang-II and increased vascular sensitivity to phenylephrine (Phe), acetylcholine, (ACh), and sodium nitroprusside (SNP). In vitro, TRV027 prevented the Ang-II-induced up-regulation of ADAM17 and in contrast with Ang-II, had no effects on ACE2 activity and expression levels. Furthermore, TRV027 induced lesser interactions between AT1R and ACE2 compared with Ang-II. Together, these data suggest that due to its biased activity for the ß-arrestin pathway, TRV027 has beneficial effects within the CNS on hypertension, autonomic and vascular function, possibly through preserving ACE2 compensatory activity in neurones.


Asunto(s)
Barorreflejo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Oligopéptidos/farmacología , Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Barorreflejo/fisiología , Presión Sanguínea/fisiología , Células HEK293 , Humanos , Hipertensión/fisiopatología , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiología , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica/efectos de los fármacos , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/metabolismo , Vasoconstrictores/farmacología
3.
J Pharmacol Exp Ther ; 356(1): 20-31, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26462539

RESUMEN

The purpose of this study was to determine whether chronic administration of Δ(9)-tetrahydrocannabinol (THC) during adolescence would (1) modify any sex-specific effects of THC on learning and (2) affect the development of tolerance to THC as an adult. Male and female rats received daily injections of saline or 5.6 mg/kg of THC from postnatal day 35-75, yielding four groups (female/saline, female/THC, male/saline, and male/THC). Rats were then trained on a procedure that assayed both learning and performance behavior and administered 0.32-18 mg/kg of THC acutely as adults (experiment 1). THC produced rate-decreasing and error-increasing effects in both sexes; however, female rats were more sensitive than male rats were to the rate-decreasing effects. Rats were then chronically administered 10 mg/kg of THC (experiment 2). Rats that received THC during adolescence developed tolerance to the rate-decreasing effects more slowly and less completely than did rats that received saline; in addition, females developed tolerance to the error-increasing effects of THC slower than males did. Western blot analysis of brain tissue indicated long-term changes in hippocampal and striatal cannabinoid type-1 receptor (CB1R) levels despite levels that were indistinguishable immediately after chronic treatment during adolescence. Striatal CB1R levels were increased in adult rats that received THC during adolescence; hippocampal CB1R levels varied by sex. In summary, female rats were more sensitive than male rats were to the acute and chronic effects of THC, and chronic administration of THC during adolescence produced long-term changes in CB1R levels that correlated with decreased tolerance development to the rate-decreasing effects of THC.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dronabinol/farmacología , Aprendizaje/efectos de los fármacos , Receptor Cannabinoide CB1/biosíntesis , Envejecimiento/psicología , Animales , Peso Corporal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Long-Evans , Receptor Cannabinoide CB1/efectos de los fármacos , Caracteres Sexuales
4.
Mol Pharmacol ; 87(5): 792-802, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25680754

RESUMEN

The human α2C-adrenergic receptor (α2C-AR) is localized intracellularly at physiologic temperature. Decreasing the environmental temperature strongly stimulates the receptor transport to the cell surface. In contrast, rat and mouse α2C-AR plasma membrane levels are less sensitive to decrease in temperature, whereas the opossum α2C-AR cell surface levels are not changed in these conditions. Structural analysis demonstrated that human α2C-AR has a high number of arginine residues in the third intracellular loop and in the C-terminus, organized as putative RXR motifs. Although these motifs do not affect the receptor subcellular localization at 37°C, deletion of the arginine clusters significantly enhanced receptor plasma membrane levels at reduced temperature. We found that this exaggerated transport of the human receptor is mediated by two functional arginine clusters, one in the third intracellular loop and one in the C-terminus. This effect is mediated by interactions with COPI vesicles, but not by 14-3-3 proteins. In rat α2C-AR, the arginine cluster from the third intracellular loop is shifted to the left due to three missing residues. Reinsertion of these residues in the rat α2C-AR restored the same temperature sensitivity as in the human receptor. Proteomic and coimmunoprecipitation experiments identified pontin as a molecule having stronger interactions with human α2C-AR compared with rat α2C-AR. Inhibition of pontin activity enhanced human receptor plasma membrane levels and signaling at 37°C. Our results demonstrate that human α2C-AR has a unique temperature-sensitive traffic pattern within the G protein-coupled receptor class due to interactions with different molecular chaperones, mediated in part by strict spatial localization of specific arginine residues.


Asunto(s)
Transporte de Proteínas/fisiología , Receptores Adrenérgicos alfa 2/metabolismo , Animales , Arginina/metabolismo , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Humanos , Proteómica/métodos , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Temperatura
5.
Am J Physiol Heart Circ Physiol ; 309(5): H926-34, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26254330

RESUMEN

We previously reported that type 2 angiotensin-converting enzyme (ACE2) compensatory activity is impaired by the disintegrin and metalloprotease 17 (ADAM17), and lack of ACE2 is associated with oxidative stress in neurogenic hypertension. To investigate the relationship between ADAM17 and oxidative stress, Neuro2A cells were treated with ANG II (100 nM) 24 h after vehicle or α-lipoic acid (LA, 500 µM). ADAM17 expression was increased by ANG II (120.5 ± 9.1 vs. 100.2 ± 0.8%, P < 0.05) and decreased after LA (69.0 ± 0.3 vs. 120.5 ± 9.1%, P < 0.05). In another set of experiments, LA reduced ADAM17 (92.9 ± 5.3 vs. 100.0 ± 11.2%, P < 0.05) following its overexpression. Moreover, ADAM17 activity was reduced by LA in ADAM17-overexpressing cells [109.5 ± 19.8 vs. 158.0 ± 20.0 fluorescence units (FU)·min(-1)·µg protein(-1), P < 0.05], in which ADAM17 overexpression increased oxidative stress (114.1 ± 2.5 vs. 101.0 ± 1.0%, P < 0.05). Conversely, LA-treated cells attenuated ADAM17 overexpression-induced oxidative stress (76.0 ± 9.1 vs. 114.1 ± 2.5%, P < 0.05). In deoxycorticosterone acetate (DOCA)-salt hypertensive mice, a model in which ADAM17 expression and activity are increased, hypertension was blunted by pretreatment with LA (119.0 ± 2.4 vs. 131.4 ± 2.2 mmHg, P < 0.05). In addition, LA improved dysautonomia and baroreflex sensitivity. Furthermore, LA blunted the increase in NADPH oxidase subunit expression, as well as the increase in ADAM17 and decrease in ACE2 activity in the hypothalamus of DOCA-salt hypertensive mice. Taken together, these data suggest that LA might preserve ACE2 compensatory activity by breaking the feedforward cycle between ADAM17 and oxidative stress, resulting in a reduction of neurogenic hypertension.


Asunto(s)
Proteínas ADAM/metabolismo , Antioxidantes/farmacología , Hipertensión/metabolismo , Estrés Oxidativo , Ácido Tióctico/farmacología , Proteínas ADAM/genética , Proteína ADAM17 , Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Antioxidantes/uso terapéutico , Barorreflejo , Línea Celular Tumoral , Hipertensión/tratamiento farmacológico , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Ácido Tióctico/uso terapéutico
6.
Hypertension ; 81(7): 1438-1449, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38567498

RESUMEN

ACE2 (angiotensin-converting enzyme 2), a multifunctional transmembrane protein, is well recognized as an important member of the (RAS) renin-angiotensin system with important roles in the regulation of cardiovascular function by opposing the harmful effects of Ang-II (angiotensin II) and AT1R (Ang-II type 1 receptor) activation. More recently, ACE2 was found to be the entry point for the SARS-CoV-2 virus into cells, causing COVID-19. This finding has led to an exponential rise in the number of publications focused on ACE2, albeit these studies often have opposite objectives to the preservation of ACE2 in cardiovascular regulation. However, notwithstanding accumulating data of the role of ACE2 in the generation of angiotensin-(1-7) and SARS-CoV-2 internalization, numerous other putative roles of this enzyme remain less investigated and not yet characterized. Currently, no drug modulating ACE2 function or expression is available in the clinic, and the development of new pharmacological tools should attempt targeting each step of the lifespan of the protein from synthesis to degradation. The present review expands on our presentation during the 2023 Lewis K. Dahl Memorial Lecture Sponsored by the American Heart Association Council on Hypertension. We provide a critical summary of the current knowledge of the mechanisms controlling ACE2 internalization and intracellular trafficking, the mutual regulation with GPCRs (G-protein-coupled receptors) and other proteins, and posttranslational modifications. A major focus is on ubiquitination which has become a critical step in the modulation of ACE2 cellular levels.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Hipertensión , Procesamiento Proteico-Postraduccional , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/metabolismo , Humanos , COVID-19/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Sistema Renina-Angiotensina/fisiología
7.
medRxiv ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38826318

RESUMEN

Background: Angiotensin (Ang)-II impairs the function of the antihypertensive enzyme ACE2 by promoting its internalization, ubiquitination and degradation thus contributing to hypertension. However, few ACE2 ubiquitination partners have been identified and their role in hypertension remains unknown. Methods: Proteomics and bioinformatic analysis were used to identify ACE2 ubiquitination partners in the brain, heart, and kidney from Ang-II-infused C57BL6/J mice from both sexes and validated the interaction between UBR1 and ACE2 in cells. Central and peripheral UBR1 knockdown was then performed in male mice to investigate its role in the maintenance of hypertension. Results: Proteomics analysis from hypothalamus identified UBR1 as a potential E3 ligase promoting ACE2 ubiquitination. Enhanced UBR1 expression, associated with ACE2 reduction, was confirmed in various tissues from hypertensive male mice and human samples. Treatment of endothelial and smooth muscle cells with testosterone, but not 17ß-estradiol, confirmed a sex-specific regulation of UBR1. In vivo silencing of UBR1 using chronic administration of small interference RNA resulted in the restoration of ACE2 levels in hypertensive males. A transient decrease in blood pressure following intracerebroventricular, but not systemic, infusion was also observed. Interestingly, UBR1 knockdown increased the brain activation of Nedd4-2, an E3 ligase promoting ACE2 ubiquitination and reduced expression of SGK1, the kinase inactivating Nedd4-2. Conclusions: These data demonstrate that UBR1 is a novel ubiquitin ligase targeting ACE2 in hypertension. UBR1 and Nedd4-2 E3 ligases appear to work synergistically to ubiquitinate ACE2. Targeting of these ubiquitin ligases may represent a novel strategy to restore ACE2 compensatory activity in hypertension.

8.
Cardiovasc Res ; 119(11): 2130-2141, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37161607

RESUMEN

AIMS: Angiotensin-converting enzyme 2 (ACE2) is a critical component of the compensatory renin-angiotensin system that is down-regulated during the development of hypertension, possibly via ubiquitination. However, little is known about the mechanisms involved in ACE2 ubiquitination in neurogenic hypertension. This study aimed at identifying ACE2 ubiquitination partners, establishing causal relationships and clinical relevance, and testing a gene therapy strategy to mitigate ACE2 ubiquitination in neurogenic hypertension. METHODS AND RESULTS: Bioinformatics and proteomics were combined to identify E3 ubiquitin ligases associated with ACE2 ubiquitination in chronically hypertensive mice. In vitro gain/loss of function experiments assessed ACE2 expression and activity to validate the interaction between ACE2 and the identified E3 ligase. Mutation experiments were further used to generate a ubiquitination-resistant ACE2 mutant (ACE2-5R). Optogenetics, blood pressure telemetry, pharmacological blockade of GABAA receptors in mice expressing ACE2-5R in the bed nucleus of the stria terminalis (BNST), and capillary western analysis were used to assess the role of ACE2 ubiquitination in neurogenic hypertension. Ubiquitination was first validated as leading to ACE2 down-regulation, and Neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2) was identified as a E3 ligase up-regulated in hypertension and promoting ACE2 ubiquitination. Mutation of lysine residues in the C-terminal of ACE2 was associated with increased activity and resistance to angiotensin (Ang)-II-mediated degradation. Mice transfected with ACE2-5R in the BNST exhibited enhanced GABAergic input to the paraventricular nucleus (PVN) and a reduction in hypertension. ACE2-5R expression was associated with reduced Nedd4-2 levels in the BNST. CONCLUSION: Our data identify Nedd4-2 as the first E3 ubiquitin ligase involved in ACE2 ubiquitination in Ang-II-mediated hypertension. We demonstrate the pivotal role of ACE2 on GABAergic neurons in the maintenance of an inhibitory tone to the PVN and the regulation of pre-sympathetic activity. These findings provide a new working model where Nedd4-2 could contribute to ACE2 ubiquitination, leading to the development of neurogenic hypertension and highlighting potential novel therapeutic strategies.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Hipertensión , Animales , Ratones , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Hipertensión/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Regulación hacia Arriba
9.
Biochim Biophys Acta ; 1813(2): 346-57, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21145921

RESUMEN

Decreasing the temperature to 30°C is accompanied by significant enhancement of α(2C)-AR plasma membrane levels in several cell lines with fibroblast phenotype, as demonstrated by radioligand binding in intact cells. No changes were observed on the effects of low-temperature after blocking receptor internalization in α(2C)-AR transfected HEK293T cells. In contrast, two pharmacological chaperones, dimethyl sulfoxide and glycerol, increased the cell surface receptor levels at 37°C, but not at 30°C. Further, at 37°C α(2C)-AR is co-localized with endoplasmic reticulum markers, but not with the lysosomal markers. Treatment with three distinct HSP90 inhibitors, radicicol, macbecin and 17-DMAG significantly enhanced α(2C)-AR cell surface levels at 37°C, but these inhibitors had no effect at 30°C. Similar results were obtained after decreasing the HSP90 cellular levels using specific siRNA. Co-immunoprecipitation experiments demonstrated that α(2C)-AR interacts with HSP90 and this interaction is decreased at 30°C. The contractile response to endogenous α(2C)-AR stimulation in rat tail artery was also enhanced at reduced temperature. Similar to HEK293T cells, HSP90 inhibition increased the α(2C)-AR contractile effects only at 37°C. Moreover, exposure to low-temperature of vascular smooth muscle cells from rat tail artery decreased the cellular levels of HSP90, but did not change HSP70 levels. These data demonstrate that exposure to low-temperature augments the α(2C)-AR transport to the plasma membrane by releasing the inhibitory activity of HSP90 on the receptor traffic, findings which may have clinical relevance for the diagnostic and treatment of Raynaud Phenomenon.


Asunto(s)
Proteínas HSP90 de Choque Térmico/fisiología , Receptores Adrenérgicos alfa 2/metabolismo , Animales , Arterias , Benzoquinonas/farmacología , Membrana Celular/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Riñón/citología , Riñón/metabolismo , Lactamas Macrocíclicas/farmacología , Macrólidos/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Transporte de Proteínas , Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Receptores Adrenérgicos alfa 2/genética , Fracciones Subcelulares , Temperatura
10.
J Neurochem ; 118(6): 1101-12, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21781118

RESUMEN

To characterize the long-term effects of adolescent marijuana abuse, we performed a proteomic analysis of cerebellar extracts from adult female rats with and without ovariectomy that were treated with Δ9-THC for 40 days during adolescence. Six proteins were found to significantly differ among the four treatment groups, with Δ9-THC and ovariectomy (OVX) decreasing the mitochondrial proteins, pyruvate carboxylase and NADH dehydrogenase, whereas the levels of putative cytosolic molecular chaperones NM23B, translationally controlled tumor protein, DJ-1 and activator of heat-shock 90kDa protein ATPase homolog 1 (AHA1) were increased. We further analyzed the effects of AHA1, a HSP90 co-chaperone, on CB1R and CB2R trafficking and signaling in transfected HEK293T and Neuro-2A cells. In HEK293T cells, AHA1 over-expression enhanced plasma membrane levels of CB1R and increased CB1R-mediated effects on cAMP levels and on MAPK phosphorylation. AHA1 over-expression also enhanced cell surface levels of endogenous CB1R and the effects of Δ9-THC on the cAMP levels in Neuro-2A cells. In contrast, over-expression of AHA1 did not affect the subcellular localization and signaling of CB2R. Our data indicate that chronic Δ9-THC administration in adolescence altered the endogenous levels of specialized proteins in the cerebellum, such as AHA1, and that this protein can change CB1R cell surface levels and signaling.


Asunto(s)
Cerebelo/metabolismo , Dronabinol/farmacología , Abuso de Marihuana/metabolismo , Chaperonas Moleculares/biosíntesis , Receptor Cannabinoide CB1/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Membrana Celular/metabolismo , Cerebelo/efectos de los fármacos , Cromatografía Líquida de Alta Presión , AMP Cíclico/metabolismo , Electroforesis en Gel Bidimensional , Femenino , Humanos , Inmunoprecipitación , Espectrometría de Masas , Microscopía Fluorescente , Chaperonas Moleculares/genética , Ovariectomía , Proteómica , Ratas , Ratas Long-Evans , Receptor Cannabinoide CB2/efectos de los fármacos , Receptores de Superficie Celular/efectos de los fármacos , Transfección
11.
Addict Biol ; 16(1): 64-81, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21158010

RESUMEN

Abuse of Δ9-THC by females during adolescence may produce long-term deficits in complex behavioral processes such as learning, and these deficits may be affected by the presence of ovarian hormones. To assess this possibility, 40 injections of saline or 5.6 mg/kg of Δ9-THC were administered i.p. daily during adolescence to gonadally intact or ovariectomized (OVX) female rats, yielding four treatment groups (intact/saline, intact/THC, OVX/saline, and OVX/ THC). Δ9-THC (0.56-10 mg/kg) was then re-administered to each of the four groups during adulthood to examine their sensitivity to its disruptive effects. The behavioral task required adult subjects to both learn (acquisition component) different response sequences and repeat a known response sequence (performance component) daily. During baseline (no injection) and control (saline injection) sessions, OVX subjects had significantly higher response rates and lower percentages of error in both behavioral components than the intact groups irrespective of saline or Δ9-THC administration during adolescence; the intact group that received Δ9-THC had the lowest response rates in each component. Upon re-administration of Δ9-THC, the groups that received adolescent ovariectomy alone, adolescent Δ9-THC administration alone, or both treatments were found to be less sensitive to the rate-decreasing effects, and more sensitive to the error-increasing effects of Δ9-THC than the control group (i.e. intact subjects that received saline during adolescence). Neurochemical analyses of the brains from each adolescent-treated group indicated that there were also persistent effects on cannabinoid type-1 (CB-1) receptor levels in the hippocampus and striatum that depended on the brain region and the presence of ovarian hormones. In addition, autoradiographic analyses of the brains from adolescent-treated, but behaviorally naïve, subjects indicated that ovariectomy and Δ9-THC administration produced effects on receptor coupling in some of the same brain regions. In summary, chronic administration of Δ9-THC during adolescence in female rats produced long-term effects on operant learning and performance tasks and on the cannabinoid system that were mediated by the presence of ovarian hormones, and that altered their sensitivity to Δ9-THC as adults.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Dronabinol/toxicidad , Estrógenos/fisiología , Alucinógenos/toxicidad , Abuso de Marihuana/fisiopatología , Progesterona/fisiología , Refuerzo en Psicología , Factores de Edad , Animales , Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Autorradiografía , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inyecciones Intraperitoneales , Ovariectomía , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas , Ratas Long-Evans , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Esquema de Refuerzo , Retención en Psicología/efectos de los fármacos , Retención en Psicología/fisiología
12.
Circ Res ; 102(6): 729-36, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18258853

RESUMEN

We recently reported the presence of angiotensin-converting enzyme (ACE)2 in brain regions controlling cardiovascular function; however, the role of ACE2 in blood pressure regulation remains unclear because of the lack of specific tools to investigate its function. We hypothesized that ACE2 could play a pivotal role in the central regulation of cardiovascular function by regulating other renin-angiotensin system components. To test this hypothesis, we generated an adenovirus expressing the human ACE2 cDNA upstream of an enhanced green fluorescent protein (eGFP) reporter gene (Ad-hACE2-eGFP). In vitro characterization shows that neuronal cells infected with Ad-hACE2-eGFP (10 to 100 multiplicities of infection), but not Ad-eGFP (100 multiplicities of infection), exhibit dose-dependent ACE2 expression and activity. In addition, an active secreted form was detected in the conditioned medium. In vivo, Ad-hACE2-eGFP infection (2x10(6) plaque-forming units intracerebroventricularly) produced time-dependent expression and activity (with a peak at 7 days) in the mouse subfornical organ. More importantly, 7 days after virus infection, the pressor response to angiotensin (Ang) II (200 pmol intracerebroventricularly) was significantly reduced in Ad-hACE2-eGFP-treated mice compared with controls. Furthermore, subfornical organ-targeted ACE2 overexpression dramatically reduced the Ang II-mediated drinking response. Interestingly, ACE2 overexpression was associated with downregulation of the Ang II type 1 receptor expression both in vitro and in vivo. These data suggest that ACE2 overexpression in the subfornical organ impairs Ang II-mediated pressor and drinking responses at least by inhibiting the Ang II type 1 receptor expression. Taken together, our results show that ACE2 plays a pivotal role in the central regulation of blood pressure and volume homeostasis, offering a new target for the treatment of hypertension and other cardiovascular diseases.


Asunto(s)
Angiotensina II/metabolismo , Barorreflejo , Conducta de Ingestión de Líquido , Neuronas/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Órgano Subfornical/metabolismo , Adenoviridae/efectos de los fármacos , Angiotensina II/administración & dosificación , Enzima Convertidora de Angiotensina 2 , Animales , Barorreflejo/efectos de los fármacos , Presión Sanguínea , Línea Celular Tumoral , Medios de Cultivo/metabolismo , Regulación hacia Abajo , Conducta de Ingestión de Líquido/efectos de los fármacos , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Frecuencia Cardíaca , Humanos , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Neuronas/enzimología , Peptidil-Dipeptidasa A/genética , Receptor de Angiotensina Tipo 1/agonistas , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Órgano Subfornical/efectos de los fármacos , Órgano Subfornical/enzimología , Factores de Tiempo , Transducción Genética , Regulación hacia Arriba
13.
Cell Signal ; 20(6): 1035-43, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18378118

RESUMEN

The molecular mechanism underlying the export of G protein-coupled receptors (GPCRs) from the endoplasmic reticulum (ER) remains largely unknown. In this manuscript, we investigated the role of Sar1 GTPase, which coordinates the assembly and budding of COPII-coated vesicles, in the cell-surface targeting, signaling and ER export of alpha(2B)-adrenergic (alpha(2B)-AR), beta(2)-AR and angiotensin II type 1 receptors (AT1R). The cell-surface expression of alpha(2B)-AR, beta(2)-AR and AT1R, and receptor-mediated ERK1/2 activation were significantly attenuated by the GTP-bound mutant Sar1H79G, suggesting that export from the ER of these receptors is mediated through the Sar1-dependent COPII-coated vesicles. Interestingly, subcellular distribution analyses showed that alpha(2B)-AR and AT1R were highly concentrated at discrete locations near the nucleus in cells expressing Sar1H79G, whereas beta(2)-AR exhibited an ER distribution. These data indicate that Sar1-catalyzed efficient GTP hydrolysis differentially regulates ER export of adrenergic and angiotensin II receptors. These data provide the first evidence indicating distinct mechanisms for the recruitment of different GPCRs into the COPII vesicles on the ER membrane.


Asunto(s)
Retículo Endoplásmico/enzimología , Proteínas de Unión al GTP Monoméricas/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Adrenérgicos/metabolismo , Línea Celular , Membrana Celular/metabolismo , Retículo Endoplásmico/química , Retículo Endoplásmico/metabolismo , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Transporte de Proteínas , Receptor de Angiotensina Tipo 1/análisis , Receptores Adrenérgicos alfa 2/análisis , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 2/análisis , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal
14.
Biochim Biophys Acta ; 1768(4): 853-70, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17074298

RESUMEN

G protein-coupled receptors (GPCRs) constitute a superfamily of cell-surface receptors which share a common topology of seven transmembrane domains and modulate a variety of cell functions through coupling to heterotrimeric G proteins by responding to a vast array of stimuli. The magnitude of cellular response elicited by a given signal is dictated by the level of GPCR expression at the plasma membrane, which is the balance of elaborately regulated endocytic and exocytic trafficking. This review will cover recent advances in understanding the molecular mechanism underlying anterograde transport of the newly synthesized GPCRs from the endoplasmic reticulum (ER) through the Golgi to the plasma membrane. We will focus on recently identified motifs involved in GPCR exit from the ER and the Golgi, GPCR folding in the ER and the rescue of misfolded receptors from within, GPCR-interacting proteins that modulate receptor cell-surface targeting, pathways that mediate GPCR traffic, and the functional role of export in controlling GPCR signaling.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencias de Aminoácidos/genética , Animales , Pliegue de Proteína , Transporte de Proteínas , Proteínas/química , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética
15.
Methods Enzymol ; 438: 217-26, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18413251

RESUMEN

Protein transport between intracellular organelles is coordinated by Rab GTPases. As an initial approach to defining the function of Rab GTPases in cardiomyocytes, our laboratory focused on Rab1, which regulates protein transport specifically from the endoplasmic reticulum (ER) to the Golgi apparatus. Our studies have demonstrated that adenovirus-driven expression of Rab1 promotes cell growth of primary cultures of neonatal cardiomyocytes in vitro and that transgenic expression of Rab1 in the myocardium induces cardiac hypertrophy in mouse hearts in vivo. These data provide strong evidence implicating that ER-to-Golgi protein transport functions as a regulatory site for control of cardiomyocyte growth. Here we describe a sets of methods used in our laboratory to characterize the function of Rab1 GTPase in modulating cardiac myocyte growth.


Asunto(s)
Miocitos Cardíacos/fisiología , Proteínas de Unión al GTP rab1/fisiología , Adenoviridae/genética , Animales , Supervivencia Celular , Células Cultivadas , Ventrículos Cardíacos/citología , Ratones , Ratones Transgénicos , Miocitos Cardíacos/virología , Ratas , Proteínas de Unión al GTP rab1/biosíntesis
16.
Cell Signal ; 18(3): 318-27, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15961277

RESUMEN

We previously demonstrated that the alpha2B-adrenergic receptor mutant, in which the F(x)6IL motif in the membrane-proximal carboxyl terminus were mutated to alanines (alpha2B-ARm), is deficient in export from the endoplasmic reticulum (ER). In this report, we determined if alpha2B-ARm could modulate transport from the ER to the cell surface and signaling of its wild-type counterpart. Transient expression of alpha2B-ARm in HEK293T cells markedly inhibited cell-surface expression of wild-type alpha2B-AR, as measured by radioligand binding. Subcellular localization demonstrated that alpha2B-ARm trapped alpha2B-AR in the ER. The alpha2B-AR was shown to form homodimers and heterodimers with alpha2B-ARm as measured by co-immunoprecipitation of the receptors tagged with green fluorescent protein and hemagglutinin epitopes. In addition to alpha2B-AR, the transport of alpha2A-AR and alpha2C-AR to the cell surface was also inhibited by alpha2B-ARm. Furthermore, transient expression of alpha2B-ARm significantly reduced cell-surface expression of endogenous alpha2-AR in NG108-15 and HT29 cells. Consistent with its effect on alpha2-AR cell-surface expression, alpha2B-ARm attenuated alpha2A-AR- and alpha2B-AR-mediated ERK1/2 activation. These data demonstrated that the ER-retained mutant alpha2B-ARm conferred a dominant negative effect on the cell-surface expression of wild-type alpha2-AR, which is likely mediated through heterodimerization. These data indicate a crucial role of ER export in the regulation of cell-surface targeting and signaling of G protein-coupled receptors.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Mutación , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Alanina/análisis , Animales , Transporte Biológico , Línea Celular , Línea Celular Tumoral , Dimerización , Humanos , Inmunoprecipitación , Leucina/análisis , Fenilalanina/análisis , Ensayo de Unión Radioligante , Ratas , Receptores Adrenérgicos alfa 2/química , Transducción de Señal/fisiología , Transfección
17.
Cell Signal ; 17(12): 1457-65, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16014327

RESUMEN

G protein-coupled receptors (GPCRs) are a superfamily of cell-surface receptors that regulate a variety of cell functions by responding to a myriad of ligands. The magnitude of the response elicited by a ligand is dictated by the level of receptor available at the plasma membrane. GPCR expression levels at the cell surface are a balance of three highly regulated, dynamic intracellular trafficking processes, namely export, internalization and degradation. This review will cover recent advances in understanding the mechanism underlying GPCR export trafficking by focusing on specific motifs required for ER export and the role of the Ras-like Rab1 GTPase and glycosylation in regulating ER-Golgi-cell-surface transport. The manifestation of diseases due to the disruption of GPCR export is also discussed.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Humanos , Datos de Secuencia Molecular , Transporte de Proteínas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Relación Estructura-Actividad , Proteínas de Unión al GTP rab/metabolismo
19.
Prog Mol Biol Transl Sci ; 132: 245-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26055062

RESUMEN

α(2C)-Adrenergic receptor (α(2C)-AR) is the least characterized adrenergic receptor subtype and still very little is known about the intracellular traffic properties and pathophysiological roles of this receptor. α(2C)-AR has an atypical subcellular localization. At 37 °C, in the vascular smooth muscle cells and in fibroblasts, the receptor is poorly localized at the plasma membrane and accumulates inside the cell. Exposure to lower temperatures stimulates α(2C)-AR transport to the cell surface. This particular intracellular trafficking of α(2C)-AR is significant in the pathology of Raynaud phenomenon. In this brief review, I will present general information on the tissue distribution and cellular localization of α(2C)-AR. Also, I will discuss the mechanisms involved in the receptor transport by focusing on the trafficking motifs and on the molecular chaperones.


Asunto(s)
Enfermedad de Raynaud/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Membrana Celular/metabolismo , ADN Helicasas/metabolismo , Fibroblastos/metabolismo , Filaminas/metabolismo , Células HEK293 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Músculo Liso Vascular/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Enfermedad de Raynaud/fisiopatología , Temperatura , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA