Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 259
Filtrar
1.
Cell ; 180(5): 862-877.e22, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142679

RESUMEN

Using untargeted metabolomics (n = 1,162 subjects), the plasma metabolite (m/z = 265.1188) phenylacetylglutamine (PAGln) was discovered and then shown in an independent cohort (n = 4,000 subjects) to be associated with cardiovascular disease (CVD) and incident major adverse cardiovascular events (myocardial infarction, stroke, or death). A gut microbiota-derived metabolite, PAGln, was shown to enhance platelet activation-related phenotypes and thrombosis potential in whole blood, isolated platelets, and animal models of arterial injury. Functional and genetic engineering studies with human commensals, coupled with microbial colonization of germ-free mice, showed the microbial porA gene facilitates dietary phenylalanine conversion into phenylacetic acid, with subsequent host generation of PAGln and phenylacetylglycine (PAGly) fostering platelet responsiveness and thrombosis potential. Both gain- and loss-of-function studies employing genetic and pharmacological tools reveal PAGln mediates cellular events through G-protein coupled receptors, including α2A, α2B, and ß2-adrenergic receptors. PAGln thus represents a new CVD-promoting gut microbiota-dependent metabolite that signals via adrenergic receptors.


Asunto(s)
Enfermedades Cardiovasculares/sangre , Microbioma Gastrointestinal/genética , Glutamina/análogos & derivados , Trombosis/metabolismo , Animales , Arterias/lesiones , Arterias/metabolismo , Arterias/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Plaquetas/metabolismo , Plaquetas/microbiología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/microbiología , Enfermedades Cardiovasculares/patología , Muerte Súbita Cardíaca/patología , Glutamina/sangre , Glutamina/genética , Humanos , Masculino , Metaboloma/genética , Metabolómica/métodos , Ratones , Infarto del Miocardio/sangre , Infarto del Miocardio/microbiología , Activación Plaquetaria/genética , Receptores Adrenérgicos alfa/sangre , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/sangre , Receptores Adrenérgicos beta/genética , Factores de Riesgo , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/microbiología , Accidente Cerebrovascular/patología , Trombosis/genética , Trombosis/microbiología , Trombosis/patología
2.
Genes Brain Behav ; 18(2): e12458, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29341465

RESUMEN

Purinergic receptor P2Y12 (P2Y12 ), a G protein-coupled purinergic receptor, is widely distributed in nervous system and involved in the progression of neurological diseases such as multiple sclerosis and neuropathic pain. The central noradrenergic system actively participates in a number of neurophysiological processes. Nevertheless, whether there is any direct relevance between P2Y12 and noradrenergic signal transduction remains unknown. In the present study, we tested the hypothesis that lack of P2Y12 impaired noradrenergic signal transduction in mouse brain. Our results showed that P2Y12 knockout (KO) mice exhibited increased anxiety-like behavior in the open-field test (OFT) and elevated plus maze test and displayed deficits in memory in the radial-arm maze test (RAMT) and Morris water maze test (MWMT). They also exhibited reduced locomotion in the OFT and MWMT. Moreover, loss of P2Y12 decreased the level of noradrenaline and the expression of noradrenergic α receptors, subtypes α2 (ARα2b) in mouse cerebellum and hippocampus. Meanwhile, it hampered the protein kinase A (PKA)/cAMP response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathway in these brain regions. Taken together, our results showed for the first time that P2Y12 KO altered the anxiety, memory and locomotion of mice, which was closely associated with abnormal state of noradrenergic system in the brain. The findings implicate that P2Y12 plays an indispensable role in noradrenergic signal transduction; its deficit is insufficient to limit anxiety responses or supports cognitive performance and activity.


Asunto(s)
Ansiedad/genética , Encéfalo/metabolismo , Memoria , Receptores Adrenérgicos alfa/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Animales , Encéfalo/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa/genética , Receptores Purinérgicos P2Y12/deficiencia , Receptores Purinérgicos P2Y12/genética , Transducción de Señal
3.
J Mol Neurosci ; 66(1): 59-67, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30091081

RESUMEN

The atypical antipsychotic drug iloperidone has high affinity for a wide range of neurotransmitter receptors, including serotonin and adrenoceptors. We examined the long-term effects of multiple doses of iloperidone (0.5, 1.5, or 5 mg/kg) on serotonin (5-HT) 5-HT1A, 5-HT2A receptor subtypes, and adrenoceptors α1 and α2 subtypes. Rats received daily intraperitoneal injections of different doses of iloperiodone or vehicle for 4 weeks. Receptor autoradiography quantified the levels of 5-HT and adrenoceptors in medial prefrontal cortex (MPC), dorsolateral frontal cortex (DFC), caudate putamen (CPu), nucleus accumbens (NAc), and hippocampal CA1 (HIP-CA1) and CA3 (HIP-CA3) regions. Four weeks of iloperidone treatment significantly and dose-dependently increased 5-HT1A and decreased 5-HT2A receptors in the MPC and DFC. Higher doses of iloperidone (1.5 and 5 mg/kg) increased 5-HT1A and decreased 5-HT2A receptors in HIP-CA1 and HIP-CA3 regions. In addition, repeated iloperidone treatment produced significant increases in α1- and α2-adrenoceptors in MPC, DFC, HIP-CA1, and HIP-CA3 regions. No changes in 5-HT and adrenoceptors were observed in other brain regions examined. These results suggest that long-term iloperidone treatment exerts region- and dose-specific effects on forebrain 5-HT and adrenoceptors, which may contribute to its therapeutic benefits in improving positive and negative symptoms of schizophrenia as well as maintaining a benign safety profile.


Asunto(s)
Antipsicóticos/farmacología , Corteza Cerebral/efectos de los fármacos , Isoxazoles/farmacología , Piperidinas/farmacología , Receptor de Serotonina 5-HT1A/genética , Receptor de Serotonina 5-HT2A/genética , Receptores Adrenérgicos alfa/genética , Animales , Ganglios Basales/efectos de los fármacos , Ganglios Basales/metabolismo , Corteza Cerebral/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Receptores Adrenérgicos alfa/metabolismo
4.
Theriogenology ; 119: 96-104, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-29990768

RESUMEN

Under physiological conditions, noradrenaline (NA) and adrenergic receptors (ARs) are implicated in the function of the uterus. The role of NA and the expression of ARs in the inflamed uterus is not fully understood. The aim of the present study was to determine the effect of inflammation on the levels of α1 (A, B, D)-, α2 (A, B, C)- and ß (1, 2, 3)-ARs mRNA and protein expression and the localization of these receptors in the porcine uterus. On Day 3 of the estrous cycle (Day 0 of the study), 50 ml of either saline (group SAL) or E. coli suspension (109 colony-forming units/ml, group E. coli) were injected into each uterine horn. In the control pigs (group CON), only laparotomy was performed. Eight days later, α1D-ARs mRNA (P < 0.001) and protein (P < 0.05) levels and α2A-ARs protein level (P < 0.05) were increased in the inflamed endometrium, while the α2C-ARs protein level (P < 0.001) was lowered, as compared to the SAL and CON groups. In the inflamed endometrium, ß2-ARs mRNA (P < 0.01) and protein (CON: P < 0.01, SAL: P < 0.001) expression was lower than in the other two groups, and ß1-ARs mRNA (P < 0.001) and protein (P < 0.01) expression was higher compared to the SAL group. After bacterial treatment, α2A- (P < 0.001) and α2B (P < 0.05) -ARs protein levels and ß2-ARs mRNA (CON: P < 0.01, SAL: P < 0.05) and protein (CON: P < 0.01, SAL: P < 0.05) expression in myometrium were found to be increased compared to both groups. In turn, in myometrium following E. coli infusion, the α2C-ARs protein level was lower (P < 0.01) than in the CON group. All studied receptors were present in the luminal and glandular epithelium, blood vessels and myometrial muscular cells of the gilt uteri in the E. coli, SAL and CON groups. The data show that inflammation changes the ARs expression in porcine uterus, suggesting their importance in the course/consequences of uterine inflammation. Those affected ARs may constitute a therapeutic target in an inflamed uterus.


Asunto(s)
Infecciones por Escherichia coli/veterinaria , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Enfermedades de los Porcinos/metabolismo , Animales , Escherichia coli , Infecciones por Escherichia coli/metabolismo , Regulación de la Expresión Génica , Transporte de Proteínas , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Porcinos , Enfermedades de los Porcinos/microbiología
5.
J Mol Neurosci ; 64(4): 567-573, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29700767

RESUMEN

Stressful situations can change biological process in human and animal, and some of these changes may transfer to the next generations. We used a communication box to induce chronic electrical foot-shock stress in rats. Tail flick latency and formalin test were done to determine the level of pain sensation. Real-time RT-PCR was used to measure the level of spinal cord µ-opioid (MOR) and α2-adrenergic receptors (α2-AR) mRNA. We demonstrate that chronic stress can change nociception and leads to hyperalgesia. Moreover, spinal cord MOR mRNA level decreased following chronic stress. We did not observe any significant changes in the level of spinal cord α2-AR mRNA between stressed and non-stressed rats. In addition, non-stressed sons of stressed mothers showed hyperalgesia compared to the control group. They showed lesser level of MOR mRNA level in comparison to the control rats. Furthermore, stressed sons of stressed mothers illustrated more hyperalgesia than the other stressed groups. We indicate that chronic stress can reduce spinal cord MOR mRNA level and lead to hyperalgesia. Additionally, these changes can transfer to offspring.


Asunto(s)
Nocicepción , Receptores Adrenérgicos alfa/metabolismo , Receptores Opioides mu/metabolismo , Médula Espinal/metabolismo , Estrés Psicológico/metabolismo , Animales , Femenino , Masculino , Exposición Materna , Ratas , Ratas Wistar , Receptores Adrenérgicos alfa/genética , Receptores Opioides mu/genética , Estrés Psicológico/fisiopatología , Destete
6.
Brain Res Bull ; 139: 157-166, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29477836

RESUMEN

Rapid eye movement sleep (REMS) plays an important role in maintaining brain excitability by regulating noradrenaline (NA) level and Na-K ATPase activity. We showed earlier that REMS deprivation (REMSD) associated elevated NA increased neuronal, while decreased glial Na-K ATPase activity. However, our knowledge was insufficient on how the REMSD-associated effect is sustained particularly under chronic condition. Using Neuro-2a cells as a model, we investigated the molecular mechanism of NA-induced increase in mRNA expression of Na-K ATPase subunit and the enzyme activity. The cells were treated with NA in the presence or absence of either α1- or ß-adrenoceptor (AR) antagonists, Ca++-channel blocker or SERCA-inhibitor, and PKA or PKC inhibitor. We observed that NA acting on α1-AR increased Na-K ATPase activity and mRNA expression of the catalytic α1- and α3-Na-K ATPase subunits in the Neuro-2a cells. Further, PLC and PKC mediated modulation of intracellular Ca++ played a critical role in inducing the mRNA expression. On the other hand NA, acting on ß-AR up-regulated expression of the regulatory ß1-subunit of Na-K ATPase. The involvement of SP1 as well as phospho-CREB transcription factors in the NA-mediated increased expression of various subunit isoforms was established. The results of this study along with that of earlier reports support our proposed working model of NA-induced increase in mRNA expression of specific Na-K ATPase subunit leading to increased Na-K ATPase activity. The findings help us understand the molecular mechanism of NA-induced increased brain excitability, for example, upon REMSD including under chronic condition.


Asunto(s)
Norepinefrina/farmacología , Receptores Adrenérgicos alfa/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Adrenérgicos/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Calcio/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/enzimología , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Neuroblastoma/patología , Prazosina/farmacología , Propranolol/farmacología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/metabolismo , Receptores Adrenérgicos alfa/genética , Detección de Señal Psicológica/efectos de los fármacos
7.
Gene ; 624: 43-49, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28456594

RESUMEN

Adrenergic receptors play a key role in activating the sympathetic nervous system, which often accompanies with the development of myocardial infarction (MI). Here, we aimed to test the association of eight potentially functional polymorphisms in five adrenergic receptor-encoding genes with MI risk. Genotypes were available for 717 MI patients and 612 controls. There were no detectable deviations from the Hardy-Weinberg equilibrium for all study polymorphisms. Allele frequencies differed remarkably for ADRA2B D/I (P<0.001), ADRB1 Ser49Gly (P=0.002), ADRB2 Gln27Glu (P=0.005), and ADRB3 Trp64Arg (P<0.001) polymorphisms, even after the Bonferroni correction. Systolic blood pressure was significantly lower in ADRA2B II genotype carriers than in the DD genotype carriers (P=0.006), while plasma high-density lipoprotein cholesterol was significantly higher in patients carrying ADRA2B I allele and ADRB1 49Ser allele than in patients with the DD genotype and 49Gly/49Gly genotype, respectively (P=0.018 and 0.033). Overall best interaction model consisted of ADRA2B D/I, ADRB1 Ser49Gly, dyslipidemia and hypertension, with the highest testing accuracy of 0.627 and the maximal 10-fold cross-validation consistency (P=0.017). Finally, a nomogram was depicted based on four significant polymorphisms and metabolic risk factors, and it had a better predictive utility and was internally validated with a discrimination C-index of 0.723 (P<0.001). Altogether, we identified two polymorphisms, ADRA2B D/I and ADRB1 Ser49Arg, which not only altered genetic susceptibility to MI, but also impacted on blood pressure and plasma lipid changes, and their combination with metabolic risk factors constituted the overall best interaction model.


Asunto(s)
Infarto del Miocardio/genética , Polimorfismo de Nucleótido Simple , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Adulto , Anciano , Presión Sanguínea/genética , Estudios de Casos y Controles , HDL-Colesterol/sangre , HDL-Colesterol/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico
8.
Circ Res ; 120(7): 1103-1115, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28219977

RESUMEN

RATIONALE: It is unknown whether every ventricular myocyte expresses all 5 of the cardiac adrenergic receptors (ARs), ß1, ß2, ß3, α1A, and α1B. The ß1 and ß2 are thought to be the dominant myocyte ARs. OBJECTIVE: Quantify the 5 cardiac ARs in individual ventricular myocytes. METHODS AND RESULTS: We studied ventricular myocytes from wild-type mice, mice with α1A and α1B knockin reporters, and ß1 and ß2 knockout mice. Using individual isolated cells, we measured knockin reporters, mRNAs, signaling (phosphorylation of extracellular signal-regulated kinase and phospholamban), and contraction. We found that the ß1 and α1B were present in all myocytes. The α1A was present in 60%, with high levels in 20%. The ß2 and ß3 were detected in only ≈5% of myocytes, mostly in different cells. In intact heart, 30% of total ß-ARs were ß2 and 20% were ß3, both mainly in nonmyocytes. CONCLUSION: The dominant ventricular myocyte ARs present in all cells are the ß1 and α1B. The ß2 and ß3 are mostly absent in myocytes but are abundant in nonmyocytes. The α1A is in just over half of cells, but only 20% have high levels. Four distinct myocyte AR phenotypes are defined: 30% of cells with ß1 and α1B only; 60% that also have the α1A; and 5% each that also have the ß2 or ß3. The results raise cautions in experimental design, such as receptor overexpression in myocytes that do not express the AR normally. The data suggest new paradigms in cardiac adrenergic signaling mechanisms.


Asunto(s)
Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animales , Células Cultivadas , Ventrículos Cardíacos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Análisis de la Célula Individual
9.
J Physiol Biochem ; 72(4): 593-604, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27405250

RESUMEN

Adiponectin exerts vasodilatory effects. Irbesartan, an angiotensin receptor blocker, possesses partial peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist activity and increases circulating adiponectin. This study explored the effect of irbesartan alone and in combination with adiponectin on blood pressure, renal hemodynamic excretory function, and vasoactive responses to angiotensin II and adrenergic agonists in spontaneously hypertensive rat (SHR). Irbesartan was given orally (30 mg/kg/day) for 28 days and adiponectin intraperitoneally (2.5 µg/kg/day) for last 7 days. Groups of SHR received either irbesartan or adiponectin or in combination. A group of Wistar Kyoto rats (WKY) served as controls. Metabolic data and plasma samples were taken on days 0, 21, and 28. In acute studies, the renal vasoconstrictor actions of angiotensin II (ANGII), noradrenaline (NA), phenylephrine (PE), and methoxamine (ME) were determined. SHR control rats had a higher mean blood pressure than the WKY (132 ± 7 vs. 98 ± 2 mmHg), lower plasma and urinary adiponectin, creatinine clearance, urine flow rate and sodium excretion, and oxidative stress markers compared to WKY (all P < 0.05) which were progressively normalized by the individual drug treatments and to a greater extent by combined treatment. Responses to intrarenal administration of NA, PE, ME, and ANGII were larger in SHR (P < 0.05) than WKY by 20-25 %. Irbesartan enhanced (P < 0.05) responses to NA and PE, while adiponectin blunted responses to all vasoconstrictors (all P < 0.05). Combined treatment in SHR further decreased the renal vascular responses to ANGII. These findings suggest that an interactive relationship may exist between PPAR-γ, alpha adrenoceptors, and ANGII in the renal vasculature of the SHR.


Asunto(s)
Adiponectina/farmacología , Antihipertensivos/farmacología , Compuestos de Bifenilo/farmacología , Hipertensión/tratamiento farmacológico , Riñón/efectos de los fármacos , PPAR gamma/genética , Tetrazoles/farmacología , Vasodilatadores/farmacología , Administración Oral , Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Regulación de la Expresión Génica , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Inyecciones Intraperitoneales , Irbesartán , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Metoxamina/farmacología , Norepinefrina/farmacología , PPAR gamma/metabolismo , Fenilefrina/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Transducción de Señal , Vasoconstrictores/farmacología
10.
Br J Pharmacol ; 173(3): 415-28, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26493129

RESUMEN

LINKED ARTICLE: This article is commented on by Michel, M. C., pp. 429-430 of this issue. To view this commentary visit http://dx.doi.org/10.1111/bph.13379. BACKGROUND AND PURPOSE: Mirabegron is the first ß3 -adrenoceptor agonist approved for treatment of overactive bladder syndrome. This study aimed to investigate the effects of ß3 -adrenoceptor agonist mirabegron in mouse urethra. The possibility that mirabegron also exerts α1 -adrenoceptor antagonism was also tested in rat smooth muscle preparations presenting α1A - (vas deferens and prostate), α1D - (aorta) and α1B -adrenoceptors (spleen). EXPERIMENTAL APPROACH: Functional assays were carried out in mouse and rat isolated tissues. Competition assays for the specific binding of [(3) H]prazosin to membrane preparations of HEK-293 cells expressing each of the human α1 -adrenoceptors, as well as ß-adrenoceptor mRNA expression and cyclic AMP measurements in mouse urethra, were performed. KEY RESULTS: Mirabegron produced concentration-dependent urethral relaxations that were shifted to the right by the selective ß3 -adrenoceptor antagonist L-748,337 but unaffected by ß1 - and ß2 -adrenoceptor antagonists (atenolol and ICI-118,551 respectively). Mirabegron-induced relaxations were enhanced by the PDE4 inhibitor rolipram, and the agonist stimulated cAMP synthesis. Mirabegron also produced rightward shifts in urethral contractions induced by the α1 -adrenoceptor agonist phenylephrine. Schild regression analysis revealed that mirabegron behaves as a competitive antagonist of α1 -adrenoceptors in urethra, vas deferens and prostate (α1A -adrenoceptor, pA2  â‰… 5.6) and aorta (α1D -adrenoceptor, pA2  â‰… 5.4) but not in spleen (α1B -adrenoceptor). The affinities estimated for mirabegron in functional assays were consistent with those estimated in radioligand binding with human recombinant α1A - and α1D -adrenoceptors (pKi  â‰… 6.0). CONCLUSION AND IMPLICATIONS: The effects of mirabegron in urethral smooth muscle are the result of ß3 -adrenoceptor agonism together with α1A and α1D -adrenoceptor antagonism.


Asunto(s)
Acetanilidas/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Tiazoles/farmacología , Uretra/efectos de los fármacos , Aminofenoles/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Células HEK293 , Humanos , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL , Relajación Muscular/efectos de los fármacos , Relajación Muscular/fisiología , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiología , Próstata/efectos de los fármacos , Próstata/fisiología , Ratas Wistar , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/fisiología , Bazo/efectos de los fármacos , Bazo/fisiología , Sulfonamidas/farmacología , Uretra/fisiología , Conducto Deferente/efectos de los fármacos , Conducto Deferente/fisiología
11.
Naunyn Schmiedebergs Arch Pharmacol ; 389(2): 243-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26660071

RESUMEN

Several previous reports suggested that many commercially available antibodies directed against G protein-coupled receptors (GPCR) lack sufficient selectivity. Accordingly, it has been proposed that receptor antibodies should be validated by at least one of several criteria, such as testing tissues or cells after knockout or silencing of the corresponding gene. Here, we tested whether 12 commercially available antibodies directed against α-adrenergic receptor (AR) subtypes (α1A/B/D, α2A/B/C), atypical chemokine receptor 3 (ACKR3), and vasopressin receptor 1A (AVPR1A) suffice these criteria. We detected in flow cytometry experiments with human vascular smooth muscle cells that the fluorescence signals from each of these antibodies were reduced by 46 ± 10 %-91 ± 2 % in cells treated with commercially available small interfering RNA (siRNA) specific for each receptor, as compared with cells that were incubated with non-targeting siRNA. The tested antibodies included anti-ACKR3 (R&D Systems, mab42273), for which specificity has previously been demonstrated. Staining with this antibody resulted in 72 ± 5 % reduction of the fluorescence signal after ACKR3 siRNA treatment. Furthermore, staining with anti-α1A-AR (Santa Cruz, sc1477) and anti-ACKR3 (Abcam, ab38089), which have previously been reported to be non-specific, resulted in 70 ± 19 % and 80 ± 4 % loss of the fluorescence signal after α1A-AR and ACKR3 siRNA treatment, respectively. Our findings demonstrate that the tested antibodies show reasonable selectivity for their receptor target under our experimental conditions. Furthermore, our observations suggest that the selectivity of GPCR antibodies depends on the method for which the antibody is employed, the species from which cells/tissues are obtained, and on the type of specimens (cell, tissue/cell homogenate, or section) tested.


Asunto(s)
Anticuerpos/inmunología , Citometría de Flujo/métodos , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , Receptores Adrenérgicos alfa/inmunología , Receptores CXCR/inmunología , Receptores de Vasopresinas/inmunología , Anticuerpos/metabolismo , Especificidad de Anticuerpos , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Sitios de Unión de Anticuerpos , Células Cultivadas , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Unión Proteica , Interferencia de ARN , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Transfección
12.
High Blood Press Cardiovasc Prev ; 22(3): 257-64, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25986078

RESUMEN

Many drugs are nowadays available to inhibit platelet activation and aggregation, especially in patients with acute coronary syndromes and undergoing percutaneous coronary intervention with stent implantation. Primary targets are represented by enzymes or receptors involved in platelet activation. Genetic mutations in these targets contribute to the inter-individual variability in platelet responses therefore weakening the efficacy of antiplatelet agents. High on treatment platelet reactivity is a condition characterized by low levels of platelet inhibition despite the use of antiplatelet drugs. This could be responsible for re-infarction, stent-thrombosis and strokes, affecting short and long-term prognosis after coronary revascularization. So far, to test antiplatelet resistance either the assessment of platelet function or the identification of genetic carriers of poly morphisms have been pursued. Although several methods are now available to test platelet reactivity, it is still debated whether its routine assessment gives real benefits in clinical practice. The present review aims at examining current evidences on genetic polymorphisms affecting optimal platelet inhibition.


Asunto(s)
Plaquetas/efectos de los fármacos , Citocromo P-450 CYP2C19/genética , Resistencia a Medicamentos/genética , Glicoproteínas de Membrana/genética , Activación Plaquetaria/genética , Inhibidores de Agregación Plaquetaria/farmacología , Plaquetas/fisiología , Clopidogrel , Citocromo P-450 CYP2C19/metabolismo , Humanos , Mutación , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/genética , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/genética , Inhibidores de Agregación Plaquetaria/metabolismo , Polimorfismo Genético , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Ticlopidina/análogos & derivados , Ticlopidina/metabolismo , Ticlopidina/farmacología
13.
Nat Commun ; 5: 5732, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25501034

RESUMEN

The cold-induced vascular response, consisting of vasoconstriction followed by vasodilatation, is critical for protecting the cutaneous tissues against cold injury. Whilst this physiological reflex response is historic knowledge, the mechanisms involved are unclear. Here by using a murine model of local environmental cold exposure, we show that TRPA1 acts as a primary vascular cold sensor, as determined through TRPA1 pharmacological antagonism or gene deletion. The initial cold-induced vasoconstriction is mediated via TRPA1-dependent superoxide production that stimulates α2C-adrenoceptors and Rho-kinase-mediated MLC phosphorylation, downstream of TRPA1 activation. The subsequent restorative blood flow component is also dependent on TRPA1 activation being mediated by sensory nerve-derived dilator neuropeptides CGRP and substance P, and also nNOS-derived NO. The results allow a new understanding of the importance of TRPA1 in cold exposure and provide impetus for further research into developing therapeutic agents aimed at the local protection of the skin in disease and adverse climates.


Asunto(s)
Hipotermia/metabolismo , Receptores Adrenérgicos alfa/genética , Piel/irrigación sanguínea , Canales de Potencial de Receptor Transitorio/genética , Vasoconstricción/genética , Acetanilidas/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Frío/efectos adversos , Regulación de la Expresión Génica , Miembro Posterior , Hipotermia/etiología , Hipotermia/genética , Hipotermia/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosforilación , Purinas/farmacología , Receptores Adrenérgicos alfa/metabolismo , Transducción de Señal , Piel/metabolismo , Piel/patología , Sustancia P/genética , Sustancia P/metabolismo , Superóxidos/metabolismo , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/deficiencia , Vasodilatación/genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
14.
Methods Mol Biol ; 1175: 245-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25150872

RESUMEN

The combination of angiotensin-converting enzyme (ACE) inhibitors and ß-adrenergic receptor (ßAR) blockers remains the essential component of heart failure (HF) pharmacotherapy. However, individual patient responses to these pharmacotherapies vary widely. The variability in response cannot be explained entirely by clinical characteristics, and genetic variation may play a role. The purpose of this chapter is to examine the current knowledge in the field of beta-blocker and ACE inhibitor pharmacogenetics in HF. ß-blocker and ACE inhibitor pharmacogenetic studies performed in patients with HF were identified from the PubMed database from 1966 to July 2011. Thirty beta-blocker and 10 ACE inhibitor pharmacogenetic studies in patients with HF were identified.The ACE deletion variant was associated with greater survival benefit from ACE inhibitors and beta-blockers compared with the ACE insertion. Ser49 in the ß1AR, the insertion in the α2CAR, and Gln41 in G protein-coupled receptor (GPCR) kinase (GRK)-5 are associated with greater survival benefit from ß-blockers, compared with Gly49, the deletion, and Leu41, respectively. However, many of these associations have not been validated. The HF pharmacogenetic literature is still in its very early stages, but there are promising candidate genetic variants that may identify which HF patients are most likely to benefit from beta-blockers and ACE inhibitors and patients that may require additional therapies.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/genética , Farmacogenética , Antagonistas Adrenérgicos beta/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Variación Genética , Genotipo , Humanos , Peptidil-Dipeptidasa A/genética , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/genética , Resultado del Tratamiento
15.
Heart Rhythm ; 11(8): 1471-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24768611

RESUMEN

BACKGROUND: Spontaneous calcium release evoking delayed afterdepolarization is believed to cause catecholaminergic polymorphic ventricular tachycardia (CPVT), a lethal human arrhythmia provoked by exercise or emotional stress. ß-Adrenergic blockers are the drug of choice, but fail to achieve complete arrhythmia control in some patients. These individuals often require flecainide, device implantation, and/or sympathetic denervation. OBJECTIVE: To optimize the arrhythmia therapy by pharmacological inhibition of the sympathetic nervous system in the homozygous calsequestrin knockout (CASQ2(Δ/Δ)) mouse model of CPVT2. METHODS: A heart telemetry device was implanted for continuous electrocardiographic recording at rest and during provocation testing. Calcium transients and abnormal calcium release were studied in cardiomyocytes isolated from adult mice. Adrenergic receptor expression was determined by using Western blotting and confocal microscopy. RESULTS: Adult CASQ2(Δ/Δ) mice suffer from complex ventricular arrhythmia at rest and ventricular tachycardia during treadmill exercise and after epinephrine injection. ß-Adrenergic blockers, propranolol and metoprolol, attenuated arrhythmia at rest but not after stress. Reserpine had no efficacy in controlling arrhythmia. Agents with α-blocking activity, phentolamine or labetalol, abolished both exercise- and epinephrine-induced arrhythmia. In contrast, injection of α-adrenergic agonist phenylephrine reproducibly provoked ventricular tachycardia. Isolated cardiomyocytes from CASQ2(Δ/Δ) mice had delayed calcium release waves upon exposure to sympathetic agonists, which were abolished by phentolamine. Hearts of calsequestrin-mutant mice expressed more α1-adrenergic receptor than did wild type control mice (P < .05). CONCLUSION: We identified a contribution of the α-adrenergic pathway to the pathogenesis of catecholamine-induced arrhythmia. α-Blockade emerges as an effective therapy in the murine model of CPVT2 and should be tried in humans resistant to ß-blockers.


Asunto(s)
Antagonistas Adrenérgicos alfa/uso terapéutico , Calsecuestrina/genética , Taquicardia Ventricular/tratamiento farmacológico , Animales , Western Blotting , Calsecuestrina/metabolismo , Modelos Animales de Enfermedad , Electrocardiografía , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatología
16.
PLoS One ; 9(3): e92167, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24670668

RESUMEN

Preterm delivery increases the risk of inadequate systemic blood flow and hypotension, and many preterm infants fail to respond to conventional inotrope treatments. If the profile of cardiac adrenoceptor subtypes in the preterm neonate is different to that at term this may contribute to these clinical problems. This study measured mRNA expression of ß1, ß2, α1A, α2A and α2B-adrenoceptor subtypes by real time PCR in term (113d), preterm (91d) and preterm piglets (91d) exposed to maternal glucocorticoid treatment. Abundance of ß-adrenoceptor binding sites in the left ventricle was measured using saturation binding assays. Relative abundance of ß1-adrenoceptor mRNA in untreated preterm hearts was ∼50% of term abundance in both left and right ventricles (P<0.001). Trends in receptor binding site density measurements supported this observation (P = 0.07). Glucocorticoid exposure increased ß1-adrenoceptor mRNA levels in the right ventricle of preterm hearts (P = 0.008) but did not alter expression in the left ventricle (P>0.1). Relative abundance of α1A-adrenoceptor mRNA was the same in preterm and term piglet hearts (P = >0.1) but was reduced by maternal glucocorticoid treatment (P<0.01); α2A-adrenoceptor mRNA abundance was higher in untreated and glucocorticoid exposed preterm piglet hearts than in term piglets (P<0.001). There was no difference between male and female piglets in mRNA abundance of any of the genes studied. In conclusion, there is reduced mRNA abundance of ß1-adrenoceptors in the preterm pig heart. If this lower expression of ß-adrenoceptors occurs in human preterm infants, it could explain their poor cardiovascular function and their frequent failure to respond to commonly used inotropes.


Asunto(s)
Corazón/embriología , Nacimiento Prematuro/metabolismo , Receptores Adrenérgicos/metabolismo , Sus scrofa/embriología , Nacimiento a Término/metabolismo , Animales , Sitios de Unión , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Miocardio/metabolismo , Receptores Adrenérgicos/genética , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo
17.
Gen Comp Endocrinol ; 195: 116-24, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24239796

RESUMEN

Catecholamines (CAs) play key roles in mediating the physiological responses to various stresses. In the present study, the expression of CA-related genes were examined in the hemocytes of scallop Chlamys farreri under heat stress, and several immune or metabolism-related parameters were investigated after heat stress and adrenoceptor antagonist stimulation. After the scallops were cultured at 28°C, the mRNA expression level of dopa decarboxylase (CfDDC) and α-adrenoceptor (CfαAR) increased significantly (P<0.01), whereas that of monoamine oxidase (CfMAO) was down-regulated in the first 6h (P<0.05), and then up-regulated to the maximum level at 24h (P<0.01). In the hemocytes of scallops injected with adrenoceptor antagonist, the expression levels of peptidoglycan-recognition protein (CfPGRP-S1) and C-type lectin (CfLec-1) began to increase significantly at 2 and 3h post propranolol and high temperature treatment, respectively (P<0.01). While the up-regulation of heat shock protein 70 (CfHSP70) post heat stress was significantly inhibited by prazosin injection (P<0.01), and that of hexokinase (CfHK) was inhibited by both prazosin and propranolol injection (P<0.01). Moreover, the remarkable increase of relative specific activity of SOD in the hemolymph post heat stress (P<0.01) was further up-regulated early after prazosin or propranolol injection (P<0.01), while that of the relative anti-bacterial ability was down-regulated by prazosin or propranolol treatment (P<0.01). These results collectively indicated that the catecholaminergic neuroendocrine system in scallop could be activated by heat stress to release CAs, which subsequently modulated the immune response and energy metabolism via α- and ß-adrenoceptors.


Asunto(s)
Catecolaminas/metabolismo , Dopa-Decarboxilasa/genética , Calor , Pectinidae/inmunología , Estrés Fisiológico , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Dopa-Decarboxilasa/metabolismo , Hemolinfa/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Pectinidae/genética , Pectinidae/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
18.
Am J Physiol Cell Physiol ; 305(10): C1080-90, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23986203

RESUMEN

Hypoglossal motoneurons (HNs) control tongue movement and play a role in maintenance of upper airway patency. Defects in these neurons may contribute to the development of sleep apnea and other cranial motor disorders including Rett syndrome (RTT). HNs are modulated by norepinephrine (NE) through α-adrenoceptors. Although postsynaptic mechanisms are known to play a role in this effect, how NE modulates the synaptic transmissions of HNs remains poorly understood. More importantly, the NE system is defective in RTT, while how the defect affects HNs is unknown. Believing that information of NE modulation of HNs may help the understanding of RTT and the design of new therapeutical interventions to motor defects in the disease, we performed these studies in which glycinergic inhibitory postsynaptic currents and intrinsic membrane properties were examined in wild-type and Mecp2(-/Y) mice, a mouse of model of RTT. We found that activation of α1-adrenoceptor facilitated glycinergic synaptic transmission and excited HNs. These effects were mediated by both pre- and postsynaptic mechanisms. The latter effect involved an inhibition of barium-sensitive G protein-dependent K(+) currents. The pre- and postsynaptic modulations of the HNs by α1-adrenoceptors were not only retained in Mecp2-null mice but also markedly enhanced, which appears to be a compensatory mechanism for the deficiencies in NE and GABAergic synaptic transmission. The existence of the endogenous compensatory mechanism is an encouraging finding, as it may allow therapeutical modalities to alleviate motoneuronal defects in RTT.


Asunto(s)
Nervio Hipogloso/citología , Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas Motoras/fisiología , Receptores Adrenérgicos alfa/metabolismo , Animales , Membrana Celular/fisiología , Fenómenos Electrofisiológicos , Regulación de la Expresión Génica/fisiología , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Mutación , Técnicas de Placa-Clamp , Receptores Adrenérgicos alfa/genética , Sinapsis
19.
Fish Shellfish Immunol ; 35(3): 825-32, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23811406

RESUMEN

The adrenergic receptors are a class of G protein-coupled receptors, through which norepinephrine and epinephrine trigger the second messenger to modulate the immune response in immunocytes of vertebrate. In the present study, a gene coding the homologue of α-adrenergic receptor was identified from scallop Chlamys farreri (designated CfαAR). Its deduced protein comprised 318 amino acids, and contained a conserved 7tm_1 domain. After CfαAR protein was expressed in the HEK293 cells, the stimulation of octopamine, tyramine, epinephrine and isoprenaline (ß-adrenergic receptor agonist) did not change significantly the intracellular cAMP concentration, whereas the stimulation of norepinephrine and phenylephrine (α-adrenergic receptor agonist) lowered significantly the cAMP level to 0.52 and 0.84 pmol µl(-1) (P < 0.05), respectively. The CfαAR transcripts were ubiquitously detected in the tested tissues including haemocytes, adductor muscle, kidney, hepatopancreas, gill, gonad and mantle, with the highest expression in the gill. The expression level of CfαAR mRNA decreased significantly (0.21-fold, P < 0.05) at 3 h after the challenge of bacteria Vibrio anguillarum. Then, it began to increase (4.74-fold, P < 0.05) at 12 h, and reached the highest level (4.92-fold, P < 0.05) at 24 h after bacteria challenge. The addition of α-adrenergic receptor agonist to the primary scallop haemocytes repressed significantly the increase of phagocytic and antibacterial activity induced by LPS stimulation, while the induction was reverted by the addition of α-adrenergic receptor antagonist. These results collectively suggested that α-adrenergic receptor could be regulated dynamically in the transcriptional level during the immune response, and it could modulate the haemocyte phagocytic and antibacterial function through the second messenger cAMP, which might be requisite for pathogen elimination and the homeostasis maintenance in scallop.


Asunto(s)
Hemocitos/citología , Hemocitos/fisiología , Pectinidae/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Datos de Secuencia Molecular , Fagocitosis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Adrenérgicos alfa/genética , Vibrio/fisiología
20.
Mol Syst Biol ; 9: 662, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23632384

RESUMEN

In pharmacology, it is crucial to understand the complex biological responses that drugs elicit in the human organism and how well they can be inferred from model organisms. We therefore identified a large set of drug-induced transcriptional modules from genome-wide microarray data of drug-treated human cell lines and rat liver, and first characterized their conservation. Over 70% of these modules were common for multiple cell lines and 15% were conserved between the human in vitro and the rat in vivo system. We then illustrate the utility of conserved and cell-type-specific drug-induced modules by predicting and experimentally validating (i) gene functions, e.g., 10 novel regulators of cellular cholesterol homeostasis and (ii) new mechanisms of action for existing drugs, thereby providing a starting point for drug repositioning, e.g., novel cell cycle inhibitors and new modulators of α-adrenergic receptor, peroxisome proliferator-activated receptor and estrogen receptor. Taken together, the identified modules reveal the conservation of transcriptional responses towards drugs across cell types and organisms, and improve our understanding of both the molecular basis of drug action and human biology.


Asunto(s)
Reposicionamiento de Medicamentos , Redes Reguladoras de Genes/efectos de los fármacos , Genoma , Hígado/efectos de los fármacos , Farmacogenética , Transcripción Genética/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Colesterol/genética , Colesterol/metabolismo , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Humanos , Hígado/citología , Hígado/metabolismo , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Ratas , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos alfa/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Especificidad de la Especie , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...