Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Biol Eng ; 41(5): 678-689, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34483791

RESUMEN

Purpose: In early 2020, the world is amid a significant pandemic due to the novel coronavirus disease outbreak, commonly called the COVID-19. Coronavirus is a lung infection disease caused by the Severe Acute Respiratory Syndrome Coronavirus 2 virus (SARS-CoV-2). Because of its high transmission rate, it is crucial to detect cases as soon as possible to effectively control the spread of this pandemic and treat patients in the early stages. RT-PCR-based kits are the current standard kits used for COVID-19 diagnosis, but these tests take much time despite their high precision. A faster automated diagnostic tool is required for the effective screening of COVID-19. Methods: In this study, a new semi-supervised feature learning technique is proposed to screen COVID-19 patients using chest CT scans. The model proposed in this study uses a three-step architecture, consisting of a convolutional autoencoder based unsupervised feature extractor, a multi-objective genetic algorithm (MOGA) based feature selector, and a Bagging Ensemble of support vector machines based binary classifier. The proposed architecture has been designed to provide precise and robust diagnostics for binary classification (COVID vs.nonCOVID). A dataset of 1252 COVID-19 CT scan images, collected from 60 patients, has been used to train and evaluate the model. Results: The best performing classifier within 127 ms per image achieved an accuracy of 98.79%, the precision of 98.47%, area under curve of 0.998, and an F1 score of 98.85% on 497 test images. The proposed model outperforms the current state of the art COVID-19 diagnostic techniques in terms of speed and accuracy. Conclusion: The experimental results prove the superiority of the proposed methodology in comparison to existing methods.The study also comprehensively compares various feature selection techniques and highlights the importance of feature selection in medical image data problems.

2.
Int J Oral Maxillofac Surg ; 50(8): 989-993, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33358522

RESUMEN

Surgical practice during the coronavirus disease 2019 (COVID-19) pandemic has changed significantly, without supporting data. With increasing experience, a dichotomy of practice is emerging, challenging existing consensus guidelines. One such practice is elective tracheostomy. Here, we share our initial experience of head and neck cancer surgery in a COVID-19 tertiary care centre, emphasizing the evolved protocol of perioperative care when compared to pre-COVID-19 times. This was a prospective study of 21 patients with head and neck cancers undergoing surgery during the COVID-19 pandemic, compared to 193 historical controls. Changes in anaesthesia, surgery, and operating room practices were evaluated. A strict protocol was followed. One patient tested positive for COVID-19 preoperatively. There was a significant increase in pre-induction tracheostomies (28.6% vs 6.7%, P=0.005), median hospital stay (10 vs 7 days, P=0.001), and postponements of surgery (57.1% vs 27.5%, P=0.01), along with a significant decrease in flap reconstructions (33.3% vs 59.6%, P=0.03). There was no mortality and no difference in postoperative morbidity. No healthcare personnel became symptomatic for COVID-19 during this period. Tracheostomy is safe during the COVID-19 pandemic and rates have increased. Despite increased rescheduling of surgeries and longer hospital stays, definitive cancer care surgery has not been deferred and maximum patient and healthcare worker safety has been ensured.


Asunto(s)
COVID-19 , Neoplasias de Cabeza y Cuello , Neoplasias de Cabeza y Cuello/cirugía , Humanos , Pandemias , Estudios Prospectivos , SARS-CoV-2 , Traqueostomía
3.
3 Biotech ; 8(1): 17, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29270333

RESUMEN

Out of 103 microsatellite markers used for studying the genetic diversity among local landraces of Luffa species, 56 were found polymorphic, including 38 gSSR and 18 eSSR, respectively. A total of 197 amplification products were obtained. The mean number of alleles per locus was 3.52. The PIC ranged from 0.037 to 0.986, while size of amplified product ranged from 105 to 500 bp. Cucumber-derived SSRs were amplified within L. acutangula (68%), L. aegyptiaca (61.16%), and L. hermaphrodita (60.2%), with an average of 63.12% cross-transferability. The Jaccard's coefficient ranged from 0.66 to 0.97, with an average of 0.81. High genetic variability was observed for node of 1st hermaphrodite flower (6.4-17), days to 1st hermaphrodite flower (38-52.1), days to 1st fruit harvest (43-65), number of fruit per cluster (1-5.9), fruit length (3.9-25 cm), fruit weight (18.4-175 g), number of fruit per plant (20-147.5), and yield per plant (2.2-4.7 kg). Two sub-populations were identified including 21 genotypes (sub-population I) and 06 genotypes (sub-population II), these two sub-populations showed 0.608-0.395% of the ancestral relationship to each other. This study provides information for future exploration, collection, and utilization of Luffa genotypes, as well as the polymorphic markers identified could be available for the study of landmarks in linkages, genomic structures, evolutionary ecology, and marker-assisted selection (MAS) in Luffa species.

4.
J Clin Invest ; 96(1): 141-9, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7615784

RESUMEN

Nitric oxide (NO) and angiotensin II (AII) can effect vascular smooth muscle cell (SMC) proliferation. However, the effects of such agents on SMC migration, an equally important phenomenon with regard to vascular pathophysiology, have received little attention. The objectives of the present study were: (a) to determine whether NO inhibits AII-induced migration of vascular SMCs; (b) to investigate the mechanism of the interaction of NO and AII on SMC migration; and (c) to evaluate the AII receptor subtype that mediates AII-induced SMC migration. Migration of rat SMCs was evaluated using a modified Boydens Chamber (transwell inserts with gelatin-coated polycarbonate membranes, 8 microns pore size). AII stimulated SMC migration in a concentration-dependent manner, and this effect was inhibited by sodium nitroprusside (SNP) and S-nitroso-N-acetylpenicillamine (SNAP). In the presence of L-arginine, but not D-arginine, IL-1 beta, an inducer of inducible NO synthase, also inhibited AII-induced SMC migration, and this effect was prevented by the NO-synthase inhibitor, N-nitro-L-arginine methyl ester. The effects of NO donors on AII-induced SMC migration were mimicked by 8-bromo-cGMP. Also, the antimigratory effects of SNAP were partially inhibited by LY83583 (an inhibitor of soluble guanylyl cyclase) and by KT5823 (an inhibitor of cGMP-dependent protein kinase). Although 8-bromo-cAMP (cAMP) also mimicked the antimigratory effects of NO donors, the antimigratory effects of SNAP were not altered by 2',5'-dideoxyadenosine (an inhibitor of adenyl cyclase) or by (R)-p-adenosine-3',5'-cyclic phosphorothioate (an inhibitor of the cAMP-dependent protein kinase). Low concentrations of the subtype AT1-receptor antagonist CGP 48933, but not the subtype AT2-receptor antagonist CGP 42112, blocked AII-induced SMC migration. These findings indicate that (a) NO inhibits AII-induced migration of vascular SMCs; (b) the antimigratory effect of NO is mediated in part via a cGMP-dependent mechanism; and (c) AII stimulates SMC migration via an AT1 receptor.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Músculo Liso Vascular/citología , Óxido Nítrico/fisiología , Nucleótidos Cíclicos/fisiología , Receptores de Angiotensina/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Antagonistas de Receptores de Angiotensina , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Masculino , Nitroprusiato/farmacología , Ratas , Ratas Sprague-Dawley
5.
J Clin Invest ; 84(4): 1155-9, 1989 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2794052

RESUMEN

The tissue uptake of extensively plasma-bound compounds is reportedly inconsistent with the conventional free-drug hypothesis limiting transport to unbound moiety in rapid intracapillary equilibrium with bound complex. Instead, protein-mediated/cell surface enhancement of dissociation has been postulated to occur in the microvasculature. This possibility was investigated by studying the passive transport of diazepam across the blood-brain barrier. Microdialysis probes placed within the vena cava and brain cortex were used to directly compare steady-state, interstitial unbound diazepam levels in both Wistar and genetically analbuminemic rats. The absence of albumin in the latter increased the unbound fraction of diazepam by almost fivefold; however, in both groups, the ratio of unbound concentrations in brain and blood at equilibrium was equal to unity. If enhanced dissociation occurred in the microvasculature, then the unbound brain level should have been greater than that in the systemic circulation. It is probable that earlier findings suggestive of protein-mediated transport reflect a nonequilibrium phenomenon. Comparison of the extent of diazepam's in vivo binding in blood by microdialysis to that estimated in vitro using conventional equilibrium dialysis with microcells showed good agreement, thus validating a widely accepted assumption of equivalency of these two values.


Asunto(s)
Barrera Hematoencefálica , Diazepam/farmacocinética , Animales , Transporte Biológico , Diazepam/sangre , Técnicas In Vitro , Ratas , Ratas Endogámicas
6.
Arterioscler Thromb Vasc Biol ; 21(11): 1745-50, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11701460

RESUMEN

Catechol-O-methyltransferase (COMT)-mediated methylation of 2-hydroxyestradiol (endogenous estradiol metabolite) to 2-methoxyestradiol (angiogenesis inhibitor) may be responsible for the antimitogenic effects of 2-hydroxyestradiol on vascular smooth muscle cells (VSMCs). Catecholamines are also substrates for COMT, and increased levels of catecholamines are associated with vasoocclusive disorders. We hypothesize that catecholamines may abrogate the vasoprotective effects of 2-hydroxyestradiol by competing for COMT and inhibiting 2-methoxyestradiol formation. To test this hypothesis, we investigated the antimitogenic effects of 0.001 to 0.1 micromol/L of 2-hydroxyestradiol on human aortic VSMC proliferation (cell number and DNA synthesis), collagen synthesis, and migration in the presence and absence of catecholamines. Norepinephrine, epinephrine, and isoproterenol concentration-dependently abrogated the inhibitory effects of 2-hydroxyestradiol on cell number, DNA synthesis, collagen synthesis, and cell migration. These modulatory/attenuating effects of catecholamines were not abrogated in the presence of the alpha- and beta-adrenergic receptor antagonists, phentolamine mesylate and propranolol, respectively. In contrast to 2-hydroxyestradiol, the antimitogenic effects of 2-methoxyestradiol (0.1 micromol/L) were not attenuated by isoproterenol (1 micromol/L) or quercetin (competitive inhibitor of COMT, 10 micromol/L). Norepinephrine, epinephrine, and isoproterenol concentration-dependently (10 to 500 micromol/L) inhibited the metabolism of 2-hydroxyestradiol (0.25 to 2 micromol/L) to 2-methoxyestradiol, and the potency of the catecholamines to reverse 2-hydroxyestradiol-induced inhibition of VSMC proliferation, collagen synthesis, and migration was correlated with their ability to inhibit 2-methoxyestradiol formation. Our findings suggest that catecholamines within the vasculature may abrogate the anti-vaso-occlusive effects of estradiol and 2-hydroxyestradiol by blocking 2-methoxyestradiol formation.


Asunto(s)
Aorta/citología , Epinefrina/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Isoproterenol/farmacología , Músculo Liso Vascular/metabolismo , Norepinefrina/farmacología , 2-Metoxiestradiol , Antagonistas Adrenérgicos alfa/farmacología , Antagonistas Adrenérgicos beta/farmacología , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Colágeno/biosíntesis , ADN/biosíntesis , Antagonismo de Drogas , Estradiol/biosíntesis , Estradiol/metabolismo , Femenino , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fentolamina/farmacología , Propranolol/farmacología
7.
Hypertension ; 33(1 Pt 2): 190-4, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9931103

RESUMEN

-The objective of this study was to characterize the effects of exogenous, drug-induced and cAMP-adenosine pathway-derived adenosine on collagen synthesis by and hypertrophy of vascular smooth muscle cells (SMCs). Confluent vascular SMCs were stimulated with 2.5% fetal calf serum in the presence and absence of adenosine receptor agonists [adenosine, 2-chloroadenosine, N6-cyclopentyladenosine, 5'-N-ethylcarboxamidoadenosine, 5'-N-methylcarboxamidoadenosine, and 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamino adenosine], drugs that increase levels of endogenous adenosine [erythro-9-(2-hydroxy-3-nonyl) adenine, dipyridamole, and iodotubericidin], and cAMP (increases adenosine by conversion to AMP and hence to adenosine via the cAMP-adenosine pathway). Adenosine receptor agonists inhibited fetal calf serum-induced collagen and total protein synthesis (as assessed by [3H]proline and [3H]leucine incorporation, respectively) with a relative potency profile consistent with the effects being mediated by adenosine A2B receptors. Erythro-9-(2-hydroxy-3-nonyl) adenine, dipyridamole, iodotubericidin, and cAMP also inhibited collagen and total protein synthesis. The effects of 2-chloroadenosine, erythro-9-(2-hydroxy-3-nonyl) adenine, iodotubericidin, and cAMP on collagen and total protein synthesis were attenuated by KF17837 and 1,3-dipropyl-8-p-sulfophenylxanthine (selective and nonselective A2 receptor antagonists, respectively) but not by 8-cyclopentyl-1, 3-dipropylxanthine (selective A1 receptor antagonist). These studies indicate that exogenous, drug-induced and cAMP-adenosine pathway-derived adenosine inhibit vascular SMC collagen synthesis and hypertrophy via A2B receptors. Thus, exogenous A2B receptor agonists and drugs that modulate endogenous adenosine levels may protect against vasoocclusive disorders by attenuating extracellular matrix synthesis by and cellular hypertrophy of vascular SMCs. Moreover, the cAMP-adenosine pathway may protect against vascular hypertrophy.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/farmacología , Aorta Abdominal/metabolismo , Colágeno/biosíntesis , Músculo Liso Vascular/metabolismo , Biosíntesis de Proteínas , 2-Cloroadenosina/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Aorta Abdominal/citología , Aorta Abdominal/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Dipiridamol/farmacología , Cinética , Leucina/metabolismo , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Prolina/metabolismo , Ratas , Ratas Sprague-Dawley , Xantinas/farmacología
8.
Hypertension ; 28(5): 765-71, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8901821

RESUMEN

In this study we determined whether cAMP is metabolized to adenosine in vascular smooth muscle cells and whether cAMP-derived adenosine modulates vascular smooth muscle cell growth. Confluent smooth muscle cells were exposed to cAMP (0.01 to 30 mumol/L) in the presence and absence of 3-isobutyl-1-methylxanthine (IBMX, 1 mmol/L; an inhibitor of both extracellular and intracellular phosphodiesterase), alpha, beta-methyleneadenosine 5'-diphosphate (AMP-CP, 100 mumol/L; an ecto-5'-nucleotidase inhibitor), and 1,3-dipropyl-8-p-sulfophenyl-xanthine (DPSPX, 100 mumol/L; a xanthine that can inhibit extracellular phosphodiesterase) for 0 to 60 minutes. Medium was then sampled and assayed for AMP, adenosine, and inosine. cAMP increased the amount of AMP, adenosine, and inosine in the medium in a time- and concentration-dependent manner. The conversion of cAMP to adenosine and inosine was inhibited by blockade of phosphodiesterase with IBMX, of ecto-phosphodiesterase with DPSPX, and of ecto-5'-nucleotidase with AMP-CP. To evaluate the physiological relevance of cAMP-derived adenosine in vascular smooth muscle cell proliferation, we studied the inhibitory effects of cAMP (10(-4) mol/L) and 8-bromo-cAMP (10(-4) mol/L) on fetal calf serum-induced DNA synthesis ([3H]thymidine incorporation) in the presence and absence of erythro-9-(2-hydroxy-3-nonyl) adenine (EHNA, an inhibitor of adenosine deaminase), dipyridamole (a blocker of adenosine transport), KF17837 (a selective A2 adenosine receptor antagonist), and DPSPX (a nonselective adenosine receptor antagonist). cAMP inhibited DNA synthesis, and both EHNA and dipyridamole enhanced this effect. Both KF17837 and DPSPX significantly reduced the inhibitory effects of cAMP on DNA synthesis; however, they did not reduce the inhibitory effects of 8-bromo-cAMP on DNA synthesis. These results indicate that vascular smooth muscle cells metabolize cAMP to adenosine via the sequential action of ecto-phosphodiesterase and ecto-5'-nucleotidase and provide the first evidence that cAMP-derived adenosine can inhibit vascular smooth muscle cell growth. Hence, this cAMP-adenosine pathway may importantly contribute to the regulation of vascular biology.


Asunto(s)
AMP Cíclico/metabolismo , Desarrollo de Músculos , Músculo Liso Vascular/crecimiento & desarrollo , Músculo Liso Vascular/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Adenosina/fisiología , Animales , Fármacos Cardiovasculares/farmacología , Células Cultivadas , Cromatografía Líquida de Alta Presión , AMP Cíclico/fisiología , ADN/biosíntesis , Inosina/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Ratas , Ratas Sprague-Dawley
9.
Hypertension ; 31(1 Pt 2): 296-302, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9453319

RESUMEN

The main purpose of this investigation was to evaluate whether the cyclic AMP-adenosine pathway, ie, the conversion of cAMP to AMP and, hence, to adenosine, is involved in the regulation of nitric oxide (NO) synthesis by vascular smooth muscle cells (SMCs). Treatment of confluent monolayers of SMCs with adenosine, 2-chloroadenosine (stable analog of adenosine), and agents that elevate endogenous (SMC-derived) adenosine (EHNA and iodotubericidin) increased nitrite/nitrate (stable metabolites of NO) levels in the medium and enhanced the conversion of 3H-L-arginine to 3H-L-citrulline by cytosolic extracts obtained from the pretreated SMCs. The stimulatory effects of adenosine were not mimicked by low (1 to 100 nmol/L) concentrations of CGS21680, an A2A receptor agonist, or CPA, a selective A1 receptor agonist. The stimulatory effects of 2-chloroadenosine and EHNA plus iodotubericidin were significantly inhibited by KF17837, a selective A2 receptor antagonist, and by DPSPX, an A1/A2 receptor antagonist, but not by DPCPX, a selective A1 receptor antagonist. DDA (adenylyl cyclase inhibitor) and Rp-cyclic AMP (protein kinase A inhibitor) did not block the effects of adenosine on NO synthesis. Incubation of SMCs with exogenous cyclic AMP, at concentrations previously shown to elevate levels ofadenosine in the medium, also increased nitrite/nitrate levels and 3H-L-citrulline formation, and the effects of cyclic AMP on NO synthesis were blocked by DPSPX and KF17837, but not by DPCPX. These findings provide evidence that exogenous and SMC-derived adenosine induce NO synthesis via A2B receptors linked to a pathway not involving adenylyl cyclase/protein kinase A. Moreover, extracellular cyclic AMP induces NO synthesis via conversion to adenosine and activation of A2B adenosine receptors. The cyclic AMP-adenosine pathway may be importantly involved in the vascular production of NO.


Asunto(s)
Adenosina/fisiología , Aorta Torácica/fisiología , AMP Cíclico/fisiología , Músculo Liso Vascular/fisiología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/biosíntesis , Receptores Purinérgicos P1/fisiología , 2-Cloroadenosina/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Citosol/metabolismo , Didesoxiadenosina/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fenetilaminas/farmacología , Agonistas del Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Tionucleótidos/farmacología
10.
Hypertension ; 31(1 Pt 2): 516-21, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9453355

RESUMEN

Adenosine inhibits rat vascular smooth muscle cell (SMC) growth. However, the effects of adenosine on human vascular SMC proliferation and synthesis of extracellular matrix proteins, such as collagen, are unknown. The objective of this study was to characterize the effects of exogenous and endogenous (SMC-derived) adenosine on human aortic SMC proliferation and collagen synthesis. Growth-arrested SMCs were stimulated with 2.5% fetal calf serum (FCS) in the presence and absence of adenosine, 2-chloroadenosine (stable adenosine analogue), and with agents that increase endogenous adenosine levels, including erythro-9-(2-hydroxy-3-nonyl) adenine (EHNA), dipyridamole, and iodotubericidin. All of these agents inhibited in a concentration-dependent manner FCS-induced SMC proliferation as assessed by DNA synthesis (3H-thymidine incorporation) and cell counting, as well as collagen synthesis (3H-proline incorporation). EHNA, dipyridamole, and iodotubericidin increased extracellular levels of adenosine by 1.7-fold to 18-fold when added separately to SMCs, and EHNA+iodotubericidin and EHNA+iodotubericidin+dipyridamole increased extracellular adenosine levels by more than 392-fold. Both KF17837 (selective A2 antagonist) and DPSPX (A1/A2 antagonist), but not DPCPX (selective A1 antagonist), blocked the antimitogenic effects of 2-chloroadenosine, EHNA, and dipyridamole on DNA and collagen synthesis, suggesting the involvement of A2A and/or A2B, but excluding the participation of A1, receptors. The lack of effect of CGS21680 (selective A2A agonist), excluded involvement of A2A receptors and suggested a major role for A2B receptors. A comparison of the inhibitory potencies of 2-chloroadenosine, N6-cyclopentyladenosine (selective A1 agonist), NECA (A1/A2 agonist), and MECA (A1/A2 agonist) were consistent with an A2B receptor subtype mediating the inhibitory effects of adenosine on human aortic SMC proliferation. In conclusion, human aortic SMCs synthesize adenosine, and exogenous as well as endogenous (SMC-derived) adenosine inhibits SMC proliferation and collagen synthesis via activation of A2B receptors.


Asunto(s)
Adenosina/farmacología , Adenosina/fisiología , Aorta Torácica/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Receptores Purinérgicos P1/fisiología , 2-Cloroadenosina/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Adenosina/análogos & derivados , Adulto , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , División Celular/efectos de los fármacos , Células Cultivadas , Colágeno/biosíntesis , ADN/biosíntesis , Dipiridamol/farmacología , Humanos , Cinética , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Fenetilaminas/farmacología , Antagonistas de Receptores Purinérgicos P1 , Ratas , Receptor de Adenosina A2B , Timidina/metabolismo , Xantinas/farmacología
11.
Hypertension ; 31(4): 943-8, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9535419

RESUMEN

The objective of this study was to characterize the effects of exogenous and endogenous (cardiac fibroblast-derived) adenosine on [3H]proline and [3H]leucine incorporation, which are reliable markers of collagen and total protein synthesis, respectively, in rat left ventricular cardiac fibroblasts. Growth-arrested confluent cardiac fibroblast monolayers were stimulated with 2.5% fetal calf serum (FCS) in the presence and absence of adenosine, 2-chloroadenosine (stable adenosine analogue), or modulators of adenosine levels including (1) erythro-9-(2-hydroxy-3-nonyl) adenine (adenosine deaminase inhibitor), (2) dipyridamole (adenosine transport blocker), and (3) iodotubericidin (adenosine kinase inhibitor). All agents inhibited in a concentration-dependent fashion FCS-induced [3H]proline and [3H]leucine incorporation. These effects were blocked by KF17837 (selective A2 antagonist) and 1,3-dipropyl-8-(p-sulfophenyl)xanthine (A1/A2 receptor antagonist) but not by 8-cyclopentyl-1,3-dipropylxanthine (selective A1 antagonist), thus excluding the participation of A1 receptors. The lack of effect of CGS21680 (selective A2A agonist) excluded involvement of A2A receptors, thus suggesting a major role for A2B receptors. Comparisons of the inhibitory potencies of N6-cyclopentyladenosine (selective A1 agonist), 5'-N-ethylcarboxamidoadenosine (A1/A2 agonist), and 5'-N-methylcarboxamidoadenosine (A1/A2 agonist) were consistent with that of an A2B receptor subtype mediating the inhibitory effects. We conclude that adenosine inhibits FCS-induced collagen and total protein synthesis in cardiac fibroblasts via activation of A2B receptors. These studies suggest, but do not prove, that endogenous adenosine may protect against cardiac fibrosis.


Asunto(s)
Adenosina/farmacología , Fármacos Cardiovasculares/farmacología , Colágeno/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Proteínas/efectos de los fármacos , Receptores Purinérgicos P1/efectos de los fármacos , 2-Cloroadenosina/farmacología , Adenosina/fisiología , Animales , Técnicas de Cultivo de Célula , División Celular/efectos de los fármacos , Colágeno/biosíntesis , Fibroblastos/citología , Fibroblastos/metabolismo , Fibrosis/metabolismo , Ventrículos Cardíacos/citología , Masculino , Infarto del Miocardio/metabolismo , Biosíntesis de Proteínas , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A2B , Receptores Purinérgicos P1/metabolismo
12.
Hypertension ; 35(1 Pt 2): 267-72, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10642309

RESUMEN

Adenosine inhibits growth of vascular smooth muscle cells. The goals of this study were to determine which adenosine receptor subtype mediates the antimitogenic effects of adenosine and to investigate the signal transduction mechanisms involved. In rat aortic vascular smooth muscle cells, platelet-derived growth factor-BB (PDGF-BB) (25 ng/mL) stimulated DNA synthesis ([(3)H]thymidine incorporation), cellular proliferation (cell number), collagen synthesis ([(3)H]proline incorporation), total protein synthesis ([(3)H]leucine incorporation), and mitogen-activated protein (MAP) kinase activity. The adenosine receptor agonists 2-chloroadenosine and 5'-N-methylcarboxamidoadenosine, but not N(6)-cyclopentyladenosine or CGS21680, inhibited the growth effects of PDGF-BB, an agonist profile consistent with an A(2B) receptor-mediated effect. The adenosine receptor antagonists KF17837 and 1,3-dipropyl-8-p-sulfophenylxanthine, but not 8-cyclopentyl-1, 3-dipropylxanthine, blocked the growth-inhibitory effects of 2-chloroadenosine and 5'-N-methylcarboxamidoadenosine, an antagonist profile consistent with an A(2) receptor-mediated effect. Antisense, but not sense or scrambled, oligonucleotides to the A(2B) receptor stimulated basal and PDGF-induced DNA synthesis, cell proliferation, and MAP kinase activity. Moreover, the growth-inhibitory effects of 2-chloroadenosine, 5'-N-methylcarboxamidoadenosine, and erythro-9-(2-hydroxy-3-nonyl) adenine plus iodotubericidin (inhibitors of adenosine deaminase and adenosine kinase, respectively) were abolished by antisense, but not scrambled or sense, oligonucleotides to the A(2B) receptor. Our findings strongly support the hypothesis that adenosine causes inhibition of vascular smooth muscle cell growth by activating A(2B) receptors coupled to inhibition of MAP kinase activity. Pharmacological or molecular biological activation of A(2B) receptors may prevent vascular remodeling associated with hypertension, atherosclerosis, and restenosis following balloon angioplasty.


Asunto(s)
Músculo Liso Vascular/química , Músculo Liso Vascular/patología , Receptores Adrenérgicos beta 2/genética , 2-Cloroadenosina/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina Quinasa/antagonistas & inhibidores , Adenosina Quinasa/metabolismo , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Anticoagulantes/farmacología , Antihipertensivos/farmacología , Aorta Abdominal/citología , Becaplermina , División Celular/efectos de los fármacos , División Celular/fisiología , Fosfatos de Dinucleósidos/farmacología , Inhibidores Enzimáticos/farmacología , Hiperplasia , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Músculo Liso Vascular/enzimología , Oligonucleótidos Antisentido/farmacología , Fenetilaminas/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 2/metabolismo , Timidina/metabolismo , Timidina/farmacología , Tritio , Tubercidina/análogos & derivados , Tubercidina/farmacología , Vasodilatadores/farmacología , Xantinas/farmacología
13.
Hypertension ; 25(4 Pt 2): 848-53, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7721443

RESUMEN

Postmenopausal women (PMW) have an increased risk of cardiovascular disease that is attenuated by hormone replacement therapy (HRT). Inasmuch as hypertension and atherosclerosis are associated with diminished endothelium-derived nitric oxide (NO), we investigated whether HRT augments NO release in PMW. We determined serum levels of nitrite/nitrate (NO2 + NO3) at baseline and during the 6th, 12th, and 24th months of the study in two groups of PMW. One group (HRT-PMW, n = 13) received continuous transdermal administration of 17 beta-estradiol (Estraderm-TTS-50) supplemented with oral norethisterone acetate (NETA) on days 1 through 12 of each month, and the other group (control PMW, n = 13) did not receive HRT. Blood samples in the HRT-PMW group were collected without regard to whether subjects were taking NETA at the time of blood sampling. Serum NO2 + NO3 levels increased in HRT-PMW for the duration of the study, whereas serum NO2 + NO3 levels remained unchanged in control PMW. When all samples regardless of timing of collection with respect to NETA treatment were included in the statistical analysis, the change in NO2 + NO3 levels in HRT-PMW was significantly greater compared with the change in control PMW (P = .037). Likewise, when only those samples collected when estradiol-treated subjects were not taking oral NETA were included in the statistical analysis, the change in NO2 + NO3 levels in the HRT-PMW group remained significant (P = .047) compared with control PMW.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Estradiol/uso terapéutico , Óxido Nítrico/sangre , Noretindrona/análogos & derivados , Posmenopausia/sangre , Quimioterapia Combinada , Estradiol/sangre , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Nitratos/sangre , Nitritos/sangre , Noretindrona/uso terapéutico , Acetato de Noretindrona
14.
Hypertension ; 33(1 Pt 2): 177-82, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9931101

RESUMEN

-Estrogens are known to induce cardioprotective effects by inhibiting smooth muscle cell (SMC) growth and neointima formation. However, the use of estrogens as cardioprotective agents is limited by carcinogenic effects in women and feminizing effects in men. If noncarcinogenic and nonfeminizing estrogenlike compounds, such as natural phytoestrogens, afford cardioprotection, this would provide a safe method for prevention of cardiovascular disease in both men and women. Therefore, we evaluated and compared in human aortic SMCs the effects of phytoestrogens (formononetin, genistein, biochanin A, daidzein, and equol) on 2.5% fetal calf serum-induced proliferation (3H-thymidine incorporation and cell number), collagen synthesis (3H-proline incorporation), and total protein synthesis (3H-leucine incorporation) and on PDGF-BB (25 ng/mL)-induced migration (modified Boydens chambers). Moreover, the effects of phytoestrogens on PDGF-BB (25 ng/mL)-induced mitogen-activated protein kinase (MAP kinase) activity in SMCs was also studied. Phytoestrogens inhibited proliferation, collagen and total protein synthesis, migration, and MAP kinase activity in a concentration-dependent manner and in the following order of potency: biochanin A>genistein>equol>daidzein>formononetin. In conclusion, our studies provide the first evidence that in human aortic SMCs phytoestrogens inhibit mitogen-induced proliferation, migration and extracellular matrix synthesis and inhibit/downregulate MAP kinase activity. Thus, phytoestrogens may confer protective effects on the cardiovascular system by inhibiting vascular remodeling and neointima formation and may be clinically useful as a safer substitute for feminizing estrogens in preventing cardiovascular disease in both women and men.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Estrógenos no Esteroides/farmacología , Músculo Liso Vascular/efectos de los fármacos , Adulto , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , Bovinos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cromanos/farmacología , Medios de Cultivo , ADN/biosíntesis , Relación Dosis-Respuesta a Droga , Equol , Femenino , Genisteína/farmacología , Humanos , Isoflavonas/farmacología , Cinética , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Fitoestrógenos , Preparaciones de Plantas , Biosíntesis de Proteínas
15.
Hypertension ; 27(3 Pt 2): 766-73, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8613238

RESUMEN

Several endogenous factors generated within the vessel wall have been implicated in contributing to the vascular remodeling process associated with hypertension and atherosclerosis. Furthermore, substances generated by smooth muscle cells (SMCs) are known to regulate SMC proliferation in an autocrine fashion. Adenosine is a vasodilator synthesized by SMCs, and exogenous adenosine inhibits SMC proliferation. However, whether adenosine produced endogenously has antimitogenic effects is not known. Hence, we evaluated the effects of SMC-derived adenosine on 2.5% fetal calf serum-induced proliferation of rat aortic SMCs. SMC proliferation was assayed by measurement of DNA synthesis ([3H]thymidine incorporation) and cell counting. To determine the effects of endogenous adenosine on SMC proliferation, we stimulated growth-arrested SMCs with 2.5% fetal calf serum in the presence and absence of modulators of adenosine levels, including (1) erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride (EHNA; inhibits adenosine deaminase), (2) dipyridamole (blocks adenosine transport and inhibits phosphodiesterase), (3) dipyridamole plus EHNA, and (4) adenosine with or without EHNA. [3H]Thymidine incorporation and cell number were measured after 24 and 96 hours, respectively. EHNA and dipyridamole inhibited both FCS-induced DNA synthesis and cell proliferation in a concentration-dependent manner. Furthermore, extracellular (in medium) adenosine levels were significantly increased when cultured cells were treated with EHNA, and the inhibitory effects of dipyridamole as well as exogenous adenosine were enhanced in the presence of EHNA. Additionally, the inhibitory effects of dipyridamole and EHNA on DNA synthesis were significantly reduced in the presence of KF17837, an A2 adenosine receptor antagonist. These results indicate that SMC-derived adenosine can inhibit SMC proliferation. Hence, it is possible that a defect in localized adenosine synthesis within the vessel wall could contribute to vascular thickening and neointima formation.


Asunto(s)
Adenosina/metabolismo , Músculo Liso Vascular/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Adenosina/antagonistas & inhibidores , Adenosina/farmacología , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Replicación del ADN/efectos de los fármacos , Dipiridamol/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Músculo Liso Vascular/citología , Ratas , Ratas Sprague-Dawley
16.
Hypertension ; 27(3 Pt 2): 786-93, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8613241

RESUMEN

Abnormal growth of vascular smooth muscle cells (SMC) is frequently associated with hypertension and atherosclerosis, and homeostasis within a normal vessel is maintained by the balanced generation of both vasoconstrictors and vasodilators. Moreover, several endogenous vasoconstricting factors induce SMC growth, whereas several vasodilators inhibit SMC growth. Inasmuch as adenosine is a potent vasodilator, it is possible that it too could inhibit SMC growth. Hence, the effects of adenosine (10(-8) to 10(-3) mol/L), 2-chloroadenosine (a stable analogue of adenosine; 10(-8) to 10(-3) mol/L), and 8-bromo-cAMP (10(-8) to 10(-3) mol/L) on fetal calf serum (FCS; 2.5%)-induced growth of rat aortic SMC were evaluated. Growth was analyzed by assaying DNA synthesis (thymidine incorporation in SMC pulsed for 4 hours with 1 microCi/mL [3H]thymidine) and cell proliferation (change in cell number). Growth-arrested SMC were treated with 2.5% FCS in the presence and absence of adenosine, 2-chloroadenosine, or 8-bromo-cAMP for 24 hours for DNA synthesis or 4 days for cell proliferation. All three substances inhibited DNA synthesis and cell proliferation in a concentration-dependent manner. Compared with adenosine, 2-chloroadenosine was more potent in inhibiting growth. The inhibitory effects of 2-chloroadenosine were reversed by KF17837 (a specific A2 receptor antagonist) but not by DPCPX (a specific A1 receptor antagonist). Furthermore, the inhibitory effects of 2-chloroadenosine were not mimicked by CGS21680 (an A2a receptor agonist), and the effects of N6-cyclopentyladenosine (CPA; an A1 receptor agonist) were not markedly more potent than those of 2-chloroadenosine, suggesting that the inhibitory effects of adenosine are possibly mediated via A2b receptors. These studies provide evidence that adenosine inhibits SMC growth and suggest that a decrease in local levels of adenosine may initiate SMC growth and contribute to the vascular remodeling process observed in hypertension and atherosclerosis.


Asunto(s)
Adenosina/farmacología , Músculo Liso Vascular/citología , Adenosina/metabolismo , Animales , Aorta/citología , División Celular/efectos de los fármacos , Células Cultivadas , Replicación del ADN/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
17.
Hypertension ; 31(1 Pt 2): 522-8, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9453356

RESUMEN

Postmenopausal women (PMW) have increased incidence of cardiovascular disease, and estrogen substitution therapy has been shown to have cardioprotective effects. Since abnormal growth of cardiac fibroblasts (CFs) is associated with hypertension and myocardial infarction and estrogen inhibits vascular smooth muscle cell (SMC) growth, it is feasible that estrogen may attenuate cardiac remodeling by inhibiting CF growth, and this possibility was investigated by using cultured CFs. 17Beta-estradiol and progesterone, but not 17alpha-estradiol, estrone, or estriol, inhibited 2.5% FCS-induced proliferation (DNA synthesis and cell number) and collagen synthesis (3H-proline incorporation) in a concentration-dependent manner and to a similar extent in male and female CFs. Compared to 17beta-estradiol, its metabolites 2-hydroxyestradiol and 2-methoxyestradiol were more potent in inhibiting FCS-induced DNA synthesis, collagen synthesis, and cell proliferation. The inhibitory effects of 17beta-estradiol and its metabolites were enhanced in presence of progesterone and 4-hydroxytamoxifen (high-affinity estrogen receptor ligand). Moreover, like estrogens, the dietary phytoestrogens biochanin A and daidzein inhibited FCS-induced growth of CFs. In conclusion, 17beta-estradiol, its metabolites, and progesterone inhibit CF growth in a gender-independent fashion. Moreover, hormone replacement therapy using 17beta-estradiol and progesterone may protect PMW against cardiovascular disease by inhibiting CF growth and cardiac remodeling; whereas estrogens that do not inhibit CF growth may be less effective in protecting PMW against cardiovascular disease. Finally, our studies provide evidence that phytoestrogens inhibit CF growth and may be clinically useful as a substitute for feminizing estrogens in preventing cardiovascular disease in both women and men.


Asunto(s)
División Celular/efectos de los fármacos , Estradiol/farmacología , Miocardio/citología , Progesterona/farmacología , 2-Metoxiestradiol , Animales , Células Cultivadas , Colágeno/biosíntesis , ADN/biosíntesis , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Estrógenos de Catecol/farmacología , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Cinética , Masculino , Miocardio/metabolismo , Prolina/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/agonistas , Caracteres Sexuales , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
18.
J Clin Endocrinol Metab ; 82(2): 388-94, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9024224

RESUMEN

Increased incidence of cardiovascular disease in postmenopausal women (PMW) is accompanied by ovarian dysfunction; hormone replacement therapy (HRT) can have cardioprotective effects. Because hypertension and atherosclerosis are associated with impaired release of endothelium-derived nitric oxide (NO) and increased levels of low-density lipoproteins (LDL), we investigated whether HRT augments NO release, and whether these increases are accompanied by a decrease in LDL levels in PMW. We determined serum nitrite/ nitrate (NO2-/NO3-) and LDL levels at baseline (before initiation of HRT) and during the 6th and 12th months of the study. The PMW (n = 26) received continuous oral administration of estradiol valerate (Progynova, 2 mg daily) for 21 days supplemented with either oral cyproterone acetate (CPA; 1 mg; n = 11) or medroxyprogesterone acetate (MPA; 5 mg; n = 15) on days 12-21 of each treatment cycle. Blood samples in the PMW receiving HRT were collected at times while the subjects were taking estradiol valerate alone and estradiol valerate plus CPA or MPA. Compared with the samples collected at baseline, serum NO2-/NO3- levels increased significantly from 20.1 +/- 1.58 mumol/L at baseline to 30 +/- 3.7 mumol/L (P < 0.01) in samples collected after 12 months of HRT while the PMW were not taking progestins (CPA or MPA), and to 25.4 +/- 2 mumol/L (P < 0.05) when all the samples, regardless of the treatment with CPA or MPA, were included in the analysis. Moreover, > 30% increase in serum NO2-/NO3- levels were observed only in 13 (responders) out of 26 PMW substituted with estradiol valerate, suggesting that estradiol may improve endogenous NO synthesis in a differential fashion. Compared with baseline, no significant increases in serum NO2-/NO3- were observed in samples collected while the estradiol-treated responders were taking either CPA or MPA. In contrast to NO2-/NO3- serum LDL levels were significantly reduced in samples collected after 12 months of HRT (P < 0.05 vs. baseline). Furthermore, levels of NO2-/NO3 showed a significant negative correlation with the levels of LDL (r2 = 0.17; P < 0.05) in the responders but not in nonresponders. These results indicate that oral administration of estradiol valerate in PMW for HRT increases circulating NO levels, an effect that may contribute to the cardioprotective effects of HRT in PMW. In addition, our data suggests but does not prove that concomitant administration of a progestin may attenuate the beneficial effects of estrogen replacement therapy with regard to NO release. Finally, our data provides evidence for the existence of responders and nonresponders to postmenopausal estrogen treatment with respect to improvement of endogenous NO levels, suggesting that a significant number, but not all, of the hormonally substituted PMW profit fully from the beneficial properties of a HRT.


Asunto(s)
Estradiol/análogos & derivados , Terapia de Reemplazo de Estrógeno , Acetato de Medroxiprogesterona/uso terapéutico , Óxido Nítrico/sangre , Posmenopausia/sangre , Estradiol/sangre , Estradiol/uso terapéutico , Femenino , Humanos , Lipoproteínas LDL/sangre , Persona de Mediana Edad , Nitratos/sangre , Nitritos/sangre
19.
Hypertension ; 35(1 Pt 2): 262-6, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10642308

RESUMEN

Estradiol inhibits smooth muscle cell growth; however, the mechanisms involved remain unclear. Because estradiol stimulates cAMP synthesis and adenosine inhibits cell growth, we hypothesized that the conversion of cAMP to adenosine (ie, the cAMP-adenosine pathway) mediates in part the inhibitory effects of estradiol on vascular smooth muscle cell growth. To test this hypothesis, we examined the effects of estradiol (0.001 to 1 micromol/L) on serum-induced DNA, collagen, and total protein synthesis and cell number in the absence and presence of 1, 3-dipropyl-8-p-sulfophenylxanthine (10 nmol/L; A(1)/A(2) adenosine receptor antagonist), KF17837 (10 nmol/L; selective A(2) adenosine receptor antagonist), 8-cyclopentyl-1,3-dipropylxanthine (10 nmol/L; selective A(1) adenosine receptor antagonist), and 2', 5'-dideoxyadenosine (10 micromol/L; adenylyl cyclase inhibitor). Estradiol inhibited all measures of cell growth, and the concentration-dependent inhibitory curves for estradiol were shifted to the right (P<0.05) by 1,3-dipropyl-8-p-sulfophenylxanthine, KF17837, and 2',5'-dideoxyadenosine but not by 8-cyclopentyl-1, 3-dipropylxanthine. Moreover, the inhibitory effects of estradiol were enhanced by stimulation of adenylyl cyclase with forskolin and by inhibition of adenosine metabolism with erythro-9-(2-hydroxy-3-nonyl)adenine plus iodotubericidin (adenosine deaminase and kinase inhibitors, respectively). Estradiol also increased levels of cAMP and adenosine, and these effects were blocked by 2',5'-dideoxyadenosine (P<0.05). Our results support the hypothesis that estradiol stimulates cAMP synthesis and cAMP-derived adenosine regulates smooth muscle cell growth via A(2) adenosine receptors. Thus, the cAMP-adenosine pathway may contribute importantly to the antivasooclusive effects of estradiol.


Asunto(s)
Adenosina/metabolismo , AMP Cíclico/metabolismo , Estradiol/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Adenilil Ciclasas/metabolismo , Animales , Antihipertensivos/farmacología , Aorta/citología , División Celular/efectos de los fármacos , Células Cultivadas , Colforsina/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , DDT/análogos & derivados , DDT/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas Fetales/farmacología , Masculino , Músculo Liso Vascular/química , Prolina/metabolismo , Prolina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P1/metabolismo , Tionucleótidos/farmacología , Timidina/metabolismo , Timidina/farmacología , Tritio , Xantinas/farmacología
20.
J Hypertens ; 14(1): 19-29, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12013490

RESUMEN

OBJECTIVE: To describe several emerging concepts regarding the biosynthesis, metabolism and biological roles of adenosine and to illustrate the possible significance of these ideas to vascular biology by proposing a hypothesis pertaining to the role of erythrocyte-derived adenosine deaminase in vaso-occlusive diseases associated with damaged erythrocytes. MULTIPLE PATHWAYS FOR ADENOSINE FORMATION: Three pathways of adenosine biosynthesis have been well established: the intracellular ATP pathway, the extracellular ATP pathway and the transmethylation pathway. A fourth pathway that has received relatively little attention, but could be particularly important in vascular smooth muscle, is the cyclic AMP-adenosine pathway. This pathway involves the extracellular or intracellular metabolism, or both, of cyclic AMP to AMP and hence to adenosine. Our recent experiments in cultured vascular smooth muscle cells, isolated vascular beds and intact animals support the existence of the cyclic AMP-adenosine pathway. Together these four pathways of adenosine formation should assure pharmacologically active levels of vascular adenosine. ANTIVASO-OCCLUSIVE ACTIONS OF ADENOSINE: The overall pharmacologic profile of adenosine suggests that this nucleoside functions to protect vascular beds from vaso-occlusive events. In this regard, some well known effects of adenosine include vasodilation, release of nitric oxide from vascular endothelial cells and inhibition of platelet aggregation, platelet adhesion, neutrophil-endothelial interactions, renin release and noradrenergic neurotransmission. Moreover, studies indicate that adenosine also releases nitric oxide from cultured vascular smooth muscle cells and inhibits vascular smooth muscle cell growth. Thus, any condition that reduces the levels of adenosine in the blood vessel wall or blood vessel-blood interface might predispose toward vaso-occlusive events. DAMAGE TO ERYTHROCYTES REDUCES ADENOSINE LEVELS: Adenosine deaminase rapidly metabolizes adenosine to inosine, which lacks antivaso-occlusive properties. Because erythrocytes are generously endowed with adenosine deaminase, any condition that damages erythrocytes will cause leakage of adenosine deaminase from erythrocytes directly onto the blood vessel wall, thus diminishing local vascular levels of adenosine. Experiments using dimethyl sulfoxide-induced hemolysis have confirmed the hypothesis that erythrocyte-derived adenosine deaminase can reduce adenosine levels in vivo. HYPOTHESIS: THE ROLE OF ERYTHROCYTE-DERIVED ADENOSINE DEAMINASE IN VASO-OCCLUSIVE DISEASES: Because multiple biosynthetic pathways maintain pharmacologically active levels of adenosine within the blood vessel wall and adenosine exerts a number of antivaso-occlusive effects, release of adenosine deaminase from erythrocytes may increase the risk for vaso-occlusive events.


Asunto(s)
Adenosina Desaminasa/fisiología , Arteriopatías Oclusivas/fisiopatología , Enfermedades Vasculares/fisiopatología , Adenosina Desaminasa/farmacología , Animales , Arteriopatías Oclusivas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA