Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Br J Pharmacol ; 137(4): 449-58, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12359626

RESUMEN

1. Human urotensin-II (hU-II), a cyclic undecapeptide, is amongst the most potent mammalian vasoconstrictors identified, suggesting that hU-II and its G-protein-coupled receptor (UT) may regulate cardiovascular homeostasis. Such a hypothesis would benefit greatly from the development of selective UT antagonists. 2. Although the somatostatin (SST) antagonist SB-710411 (Cpa-c[D-Cys-Pal-D-Trp-Lys-Val-Cys]-Cpa-amide) is purported to block U-II-induced contractions in rat isolated aorta, little is known about its specific pharmacological properties. 3. SB-710411 (10 micro M) inhibited hU-II-induced contraction in rat isolated aorta causing a significant, parallel shift in the agonist concentration-response curve (pK(b) 6.28+/-0.11; n=8) with no suppression of the E(max). In contrast, SB-710411 did not alter the contractile actions of angiotensin-II, phenylephrine, or KCl. Paradoxically, however, SB-710411 potentiated the contractile response to endothelin-1 (pEC(50) 8.02+/-0.16 and 8.54+/-0.11, P<0.01; n=8). Rather than being specific toSB-710411, this phenomenon appears to be related to somatostatin receptor affinity and not intrinsic activity since the SST agonist somatostatin-14 and antagonist cyclo-somatostatin also potentiated endothelin-1-induced contraction. 4. SB-710411 (10 micro M) did not inhibit carbachol, sodium nitroprusside, IBMX, isoprenaline, and levcromakalim-induced reversal of tone established with noradrenaline. In contrast, however, SB-710411 significantly inhibited the reversal of tone established with endothelin-1 using the same vasorelaxants. 5. In summary, although SB-710411 inhibits the vasoconstrictor actions of hU-II in a competitive, surmountable manner, it also possesses additional pharmacological actions. Thus, whilst the present study is amongst the first to detail the properties of a functional U-II receptor antagonist, the data suggest caution be used when assessing data generated utilizing this moiety and other SST analogues.


Asunto(s)
Péptidos Cíclicos/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores Acoplados a Proteínas G , Urotensinas/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/fisiología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología
2.
Br J Pharmacol ; 139(2): 203-7, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12770925

RESUMEN

The functional activity of the peptidic neuromedin B receptor antagonist BIM-23127 was investigated at recombinant and native urotensin-II receptors (UT receptors). Human urotensin-II (hU-II) promoted intracellular calcium mobilization in HEK293 cells expressing the human UT (hUT) or rat UT (rUT) receptors with pEC(50) values of 9.80+/-0.34 (n=6) and 9.06+/-0.32 (n=4), respectively. While BIM-23127 alone had no effect on calcium responses in either cell line, it was a potent and competitive antagonist at both hUT (pA(2)=7.54+/-0.14; n=3) and rUT (pA(2)=7.70+/-0.05; n=3) receptors. Furthermore, BIM-23127 reversed hU-II-induced contractile tone in the rat-isolated aorta with a pIC(50) of 6.66+/-0.04 (n=4). In conclusion, BIM- 23127 is the first hUT receptor antagonist identified to date and should not be considered as a selective neuromedin B receptor antagonist.


Asunto(s)
Péptidos Cíclicos/farmacología , Receptores de Bombesina/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Aorta Torácica/efectos de los fármacos , Unión Competitiva , Calcio/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Urotensinas/farmacología
3.
Br J Pharmacol ; 142(6): 921-32, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15210573

RESUMEN

1 In an effort to identify endogenous, native mammalian urotensin-II (U-II) receptors (UT), a diverse range of human, primate and rodent cell lines (49 in total) were screened for the presence of detectable [125I]hU-II binding sites. 2 UT mRNA (Northern blot, PCR) and protein (immunocytochemistry) were evident in human skeletal muscle tissue and cells. 3 [(125)I]hU-II bound to a homogenous population of high-affinity, saturable (Kd 67.0+/-11.8 pm, Bmax 9687+/-843 sites cell(-1)) receptors in the skeletal muscle (rhabdomyosarcoma) cell line SJRH30. Radiolabel was characteristically slow to dissociate (< or =15% dissociation 90 min). A lower density of high-affinity U-II binding sites was also evident in the rhabdomyosarcoma cell line TE671 (1667+/-165 sites cell(-1), Kd 74+/-8 pm). 4 Consistent with the profile recorded in human recombinant UT-HEK293 cells, [125I]hU-II binding to SJRH30 cells was selectively displaced by both mammalian and fish U-II isopeptides (Kis 0.5+/-0.1-1.2+/-0.3 nm) and related analogues (hU-II[4-11]>[Cys(5,10)]Acm hU-II; Kis 0.4+/-0.1 and 864+/-193 nm, respectively). 5 U-II receptor activation was functionally coupled to phospholipase C-mediated [Ca2+]i mobilization (EC50 6.9+/-2.2 nm) in SJRH30 cells. 6 The present study is the first to identify the presence of 'endogenous' U-II receptors in SJRH30 and TE671 cells. SJRH30 cells, in particular, might prove to be of utility for (a) investigating the pharmacological properties of hU-II and related small molecule antagonists at native human UT and (b) delineating the role of this neuropeptide in the (patho)physiological regulation of mammalian neuromuscular function.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Rabdomiosarcoma/metabolismo , Animales , Sitios de Unión , Unión Competitiva/efectos de los fármacos , Northern Blotting , Péptido Relacionado con Gen de Calcitonina/farmacología , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Células HeLa , Humanos , Hormonas Hipotalámicas/farmacología , Inmunohistoquímica , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Radioisótopos de Yodo , Cinética , Masculino , Melaninas/farmacología , Neuropéptido Y/farmacología , Neurofisinas/farmacología , Toxina del Pertussis/farmacología , Hormonas Hipofisarias/farmacología , Precursores de Proteínas/farmacología , Ensayo de Unión Radioligante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rabdomiosarcoma/patología , Tapsigargina/farmacología , Urotensinas/genética , Urotensinas/metabolismo , Urotensinas/farmacología , Vasopresinas/farmacología
4.
Eur J Pharmacol ; 492(2-3): 113-6, 2004 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-15178353

RESUMEN

SB-710411 (Cpa-c[d-Cys-Pal-d-Trp-Lys-Val-Cys]-Cpa-amide) inhibited [(125)I]urotensin-II rat and monkey UT receptor binding (pK(i)s 7.50+/-0.07 and 6.82+/-0.06). However, whereas SB-710411 antagonized urotensin-II-induced inositol phosphate formation at the rat UT receptor (pK(b) 6.54+/-0.05), this ligand functioned as an agonist at the monkey UT receptor (pEC(50) 6.56+/-0.35, E(max) 5.27+/-0.65-fold over basal). Indeed, in contrast to the rat UT receptor (and rat isolated arteries), SB-710411 exhibited intrinsic activity in monkey arteries acting as an efficacious vasoconstrictor (pEC(50)s 5.03+/-0.18 to 5.71+/-0.21, E(max)s 101+/-4 to 218+/-58% KCl). These data demonstrate that caution must be taken when extrapolating the pharmacology of a specific ligand(s) between the rodent and primate UT receptors.


Asunto(s)
Péptidos Cíclicos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Urotensinas/farmacología , Vasoconstrictores/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aorta Torácica/fisiología , Arterias/efectos de los fármacos , Arterias/fisiología , Línea Celular , Humanos , Técnicas In Vitro , Fosfatos de Inositol/biosíntesis , Ligandos , Macaca fascicularis , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Ensayo de Unión Radioligante , Ratas , Especificidad de la Especie
5.
Naunyn Schmiedebergs Arch Pharmacol ; 370(4): 238-50, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15549273

RESUMEN

Urotensin-II (U-II), the most potent mammalian vasoconstrictor identified, and its receptor, UT, exhibits increased expression in cardiac tissue and plasma in congestive heart failure (CHF) patients. Cardiomyocyte hypertrophy is primarily responsible for increased myocardial mass associated with cardiac injury. Neurohumoral factors such as angiotensin-II, endothelin-1, catecholamines, and inflammatory cytokines are thought to mediate this response. U-II shares similar biological activities with other hypertrophic G(q)-coupled receptor ligands such as angiotensin-II and endothelin-1, but a role for U-II in cardiomyocyte hypertrophy has not been characterized. The hypothesis of the current study was that U-II, acting through its G(q)-coupled receptor UT plays a hypertrophic role in cardiac hypertrophic remodeling. We report that adenoviral upregulation of the UT receptor "unmasked" U-II-induced hypertrophy in H9c2 cardiomyocytes, with a threshold response of 202+/-8 binding sites/cell. U-II was equally as efficacious as phenylephrine in inducing hypertrophy, measured by a reporter assay (EC(50) 0.7+/-0.2 nM) and [(3)H]-leucine incorporation (EC(50) 150+/-40 nM). A competitive peptidic UT receptor antagonist, BIM-23127, inhibited U-II-induced hypertrophy ( K(B) 34+/-6 nM). U-II did not affect cell proliferation or apoptosis, indicating that U-II is more hypertrophic than apoptotic or hyperplastic in cardiomyocytes. U-II (10 nM) stimulated interleukin-6 release in UT-expressing cardiomyocytes (4.6-fold at 6 h). Finally, in a rat heart failure model, cardiac ventricular mRNA expression of U-II, UT receptor, interleukin-6, and interleukin-1-beta is increased time-dependently following myocardial injury. These results indicate that U-II might play a role in cardiac remodeling associated with CHF by stimulation of cardiomyocyte hypertrophy via UT, and through upregulation of inflammatory cytokines. As such, UT antagonism may represent a novel therapeutic target for the clinical management of heart failure.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/patología , Mediadores de Inflamación/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Urotensinas/farmacología , Animales , Línea Celular , Citocinas/biosíntesis , Citocinas/fisiología , Relación Dosis-Respuesta a Droga , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Masculino , Miocitos Cardíacos/metabolismo , Ratas , Ratas Endogámicas Lew , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/biosíntesis
6.
J Biol Chem ; 279(12): 11456-64, 2004 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-14670966

RESUMEN

UTP is a potent full agonist at both the human P2Y(4) (hP2Y(4)) and rat P2Y(4) (rP2Y(4)) receptor. In contrast, ATP is a potent full agonist at the rP2Y(4) receptor but is a similarly potent competitive antagonist at the hP2Y(4) receptor. To delineate the structural determinants of agonism versus antagonism in these species homologues, we expressed a series of human/rat P2Y(4) receptor chimeras in 1321N1 human astrocytoma cells and assessed the capacity of ATP and UTP to mobilize intracellular Ca(2+). Replacement of the NH(2) terminus of the hP2Y(4) receptor with the corresponding region of the rP2Y(4) receptor resulted in a receptor that was activated weakly by ATP, whereas replacement of the second extracellular loop (EL2) of the hP2Y(4) receptor with that of the rP2Y(4) receptor yielded a chimeric receptor that was activated fully by UTP and near fully by ATP, albeit with lower potencies than those observed at the rP2Y(4) receptor. These potencies were increased, and ATP was converted to a full agonist by replacing both the NH(2) terminus and EL2 in the hP2Y(4) receptor with the corresponding regions from the rP2Y(4) receptor. Mutational analysis of the five divergent amino acids in EL2 between the two receptors revealed that three amino acids, Asn-177, Ile-183, and Leu-190, contribute to the capacity of EL2 to impart ATP agonism. Taken together, these results suggest that the second extracellular loop and the NH(2) terminus form a functional motif that plays a key role in determining whether ATP functions as an agonist or antagonist at mammalian P2Y(4) receptors.


Asunto(s)
Adenosina Trifosfato/farmacología , Receptores Purinérgicos P2/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Agonistas del Receptor Purinérgico P2 , Antagonistas del Receptor Purinérgico P2 , Ratas , Receptores Purinérgicos P2/química , Proteínas Recombinantes de Fusión/agonistas , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/efectos de los fármacos , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA