Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Martín Aldasoro is active.

Publication


Featured researches published by Martín Aldasoro.


British Journal of Pharmacology | 1997

Potentiation by vasopressin of adrenergic vasoconstriction in the rat isolated mesenteric artery

Inmaculada Noguera; Pascual Medina; Gloria Segarra; M.C. Martínez; Martín Aldasoro; J. M. Vila; Salvador Lluch

1 The aim of the present study was to investigate in rat mesenteric artery rings whether low concentrations of vasopressin could modify the contractile responses to noradrenaline and electrical stimulation of perivascular nerves. 2 Vasopressin (10−10–10−7 M) caused concentration‐dependent contractions (pD2=8.36±0.09). The V1‐receptor antagonist d(CH2)5Tyr(Me)AVP (10−9–10−8 M) produced parallel rightward shifts of the control curve for vasopressin. Schild analysis yielded a pA2 value of 9.83 with a slope of 1.10±0.14. 3 Vasopressin (3×10 −10 and 10−9 M) caused concentration‐dependent potentiation of the contractions elicited by electrical stimulation (2–8 Hz; 0.2 ms duration for 30 s) and produced leftward shifts of the concentration‐response curve for noradrenaline. The V1‐receptor antagonist induced concentration‐dependent inhibitions of potentiation induced by vasopressin. The selective V1‐receptor agonist [Phe*, Orn8]‐vasotocin (3×10 −10 and 10−9 M) induced potentiation of electrical stimulation‐evoked responses which was also inhibited in the presence of the V1 antagonist (10−8 M). In contrast, the V2‐receptor agonist deamino‐8‐D‐arginine vasopressin (desmopressin 10−8–10−7 M) did not modify the electrical stimulation‐induced responses and the V2‐receptor antagonist [d(CH2)5, D‐Ile*, Ile4, Arg8]‐vasopressin (10−8–10−7 M) did not affect the potentiation evoked by vasopressin. 4 In artery rings contracted by 10−6 M noradrenaline in the presence of 10−6 M guanethidine and 10−6 M atropine, electrical stimulation (2, 4 and 8 Hz) produced frequency‐dependent relaxations which were unaffected by 10−9 M vasopressin but abolished by 10−6 M tetrodotoxin. 5 Vasopressin also potentiated contractions elicited by KCl and contractions induced by addition of CaCl2 to KCl depolarized vessels. The augmenting effects were inhibited by the V1 antagonist. 6 In the presence of the calcium antagonist nifedipine (10−6 M), vasopressin failed to enhance the contractile responses to electrical stimulation, noradrenaline and KCl. 7 The results demonstrate that low concentrations of vasopressin strongly potentiate the contractions to adrenergic stimulation and KCl depolarization. This effect appears to be mediated by V1 receptor stimulation which brings about an increase in calcium entry through dihydropyridine‐sensitive calcium channels.


Stroke | 1999

Effects of Some Guanidino Compounds on Human Cerebral Arteries

Gloria Segarra; Pascual Medina; Rosa María Ballester; Paloma Lluch; Martín Aldasoro; J. M. Vila; Salvador Lluch

BACKGROUND AND PURPOSE Accumulation of endogenous guanidino-substituted analogues of L-arginine in chronic renal failure might contribute to some of the vascular and neurological disorders of this pathology. We tested the hypothesis that in human cerebral arteries, some guanidino compounds may increase vascular tone, through nitric oxide (NO) synthase inhibition, and impair endothelium-dependent relaxation. METHODS Rings of human middle cerebral artery were obtained during autopsy of 26 patients who had died 3 to 12 hours before. The rings were suspended in organ baths for isometric recording of tension. We then studied the responses to N(G)-monomethyl-L-arginine (L-NMMA), N(G),N(G)-dimethyl-L-arginine (asymmetrical dimethylarginine; ADMA), aminoguanidine (AG), and methylguanidine (MG). RESULTS L-NMMA (10(-6) to 3x10(-4) mol/L) and ADMA (10(-6) to 3x10(-4) mol/L) caused concentration- and endothelium-dependent contractions (median effective concentrations [EC(50)]=1.1x10(-5) and 1.6x10(-5) mol/L, respectively; E(max)=35. 5+/-7.9% and 43.9+/-5.9% of the response to 100 mmol/L KCl). AG (10(-5) to 3x10(-3) mol/L) and MG (10(-5) to 3x10(-3) mol/L) produced endothelium-independent contractions (E(max)=44.3+/-8.8% and 45.7+/-5.8% of the response to 100 mmol/L KCl, respectively). L-Arginine (10(-3) mol/L) prevented the contractions by L-NMMA and ADMA but did not change contractions induced by AG and MG. L-NMMA and ADMA inhibited endothelium-dependent relaxation induced by acetylcholine in a concentration-dependent manner; AG and MG were without effect. CONCLUSIONS The results suggest that the contractions induced by L-NMMA and ADMA are due to inhibition of endothelial NO synthase activity, whereas AG and MG do not affect the synthesis of NO. An increase in the plasma concentration of L-NMMA and ADMA associated with uremia is likely to represent a diminished release or effect of NO, and consequently, an increased cerebrovascular tone in uremic patients is highly conceivable.


British Journal of Pharmacology | 1994

Relaxation of human isolated mesenteric arteries by vasopressin and desmopressin

M.C. Martínez; J. M. Vila; Martín Aldasoro; Pascual Medina; Blas Flor; Salvador Lluch

1 The effects of vasopressin and deamino‐8‐D‐arginine vasopressin (DDAVP, desmopressin) were studied in artery rings (0.8‐1 mm in external diameter) obtained from portions of human omentum during the course of abdominal operations (27 patients). 2 In arterial rings under resting tension, vasopressin produced concentration‐dependent, endothelium‐independent contractions with an EC50 of 0.59 ± 0.12 nm. The V1 antagonist d(CH2)5Tyr(Me)AVP (1 μ) and the mixed V1‐V2 antagonist desGly‐d(CH2)5D‐Tyr(Et)ValAVP (0.01 μm) displaced the control curve to vasopressin to the right in a parallel manner without differences in the maximal responses. In the presence of indomethacin (1 μm) the contractile response to vasopressin was significantly increased (P < 0.01). 3 In precontracted arterial rings, previously treated with the V1 antagonist, d(CH2)5Tyr(Me)AVP (1 μm), vasopressin produced endothelium‐dependent relaxation. This relaxation was reduced significantly (P < 0.05) by indomethacin (1 μm) and unaffected by the V1‐V2 receptor antagonist desGly‐d(CH2)5D‐Tyr(Et)ValAVP (1 μm) or by NG‐nitro‐L‐arginine methyl ester (L‐NAME, O.1 μm). 4 The selective V2 receptor agonist, DDAVP, caused endothelium‐independent, concentration‐dependent relaxations in precontracted arterial rings that were inhibited by the mixed V1‐V2 receptor antagonist, but not by the V1 receptor antagonist or by pretreatment with indomethacin or L‐NAME. 5 Results from this study suggest that vasopressin is primarily a constrictor of human mesenteric arteries by V1 receptor stimulation; vasopressin causes dilatation only during V, receptor blockade. The relaxation appears to be mediated by the release of vasodilator prostaglandins from the endothelial cell layer and is independent of V2 receptor stimulation or release of nitric oxide. In contrast, the relaxation induced by DDAVP is largely dependent on stimulation of V2 receptors.


British Journal of Pharmacology | 1990

Role of endothelium and calcium channels in endothelin-induced contraction of human cerebral arteries

de Aguilera Em; Irurzun A; José M. Vila; Martín Aldasoro; Galeote Ms; Salvador Lluch

Endothelin constricted human isolated cerebral arteries in a concentration‐dependent manner. The maximal tension developed, as well as EC50 values were similar in arteries with and without endothelium. Removal of extracellular calcium or addition of the calcium antagonist nicardipine (10−6 m), attenuated but did not abolish responses to endothelin. These experiments show that the endothelin‐induced contraction in human cerebral arteries is not linked to the presence of intact endothelial cells. The data also show that the contractile effects of endothelin cannot be explained solely by an action on voltage‐dependent calcium channels.


Current Pharmaceutical Design | 2013

Endothelial Dysfunction in Morbid Obesity

María Dolores Mauricio; Martín Aldasoro; Joaquin Ortega; José M. Vila

Morbid obesity is a chronic multifunctional disease characterized by an accumulation of fat. Epidemiological studies have shown that obesity is associated with cardiovascular and metabolic disorders. Endothelial dysfunction, as defined by an imbalance between relaxing and contractile endothelial factors, plays a central role in the pathogenesis of these cardiometabolic diseases. Diminished bioavailability of nitric oxide (NO) contributes to endothelial dysfunction and impairs endothelium- dependent vasodilatation. But this is not the only mechanism that drives to endothelial dysfunction. Obesity has been associated with a chronic inflammatory process, atherosclerosis, and oxidative stress. Moreover levels of asymmetrical dimethyl-L-arginine (ADMA), an endogenous inhibitor of endothelial nitric oxide synthase (eNOS), are elevated in obesity. On the other hand, increasing prostanoid-dependent vasoconstriction and decreasing vasodilator prostanoids also lead to endothelial dysfunction in obesity. Other mechanisms related to endothelin-1 (ET-1) or endothelium derived hyperpolarizing factor (EDHF) have been proposed. Bariatric surgery (BS) is a safe and effective means to achieve significant weight loss, but its use is limited only to patients with severe obesity including morbid obesity. BS also proved efficient in endothelial dysfunction reduction improving cardiovascular and metabolic comorbidities associated with morbid obesity such as diabetes, coronary artery disease, nonalcoholic fatty liver disease and cancer. This review will provide a brief overview of the mechanisms that link obesity with endothelial dysfunction, and how weight loss is a cornerstone treatment for cardiovascular comorbidities obesity-related. A better understanding of the mechanisms of obesity-induced endothelial dysfunction may help develop new therapeutic strategies to reduce cardiovascular morbidity and mortality.


Circulation | 1998

Arginine Vasopressin Enhances Sympathetic Constriction Through the V1 Vasopressin Receptor in Human Saphenous Vein

Pascual Medina; Antonio Acuña; Juan Martínez-León; Eduardo Otero; José M. Vila; Martín Aldasoro; Salvador Lluch

BACKGROUND Arginine vasopressin (AVP) not only acts directly on blood vessels through V1 receptor stimulation but also may modulate adrenergic-mediated responses in animal experiments in vivo and in vitro. The aim of the present study was to investigate whether AVP can contribute to an abnormal adrenergic constrictor response of human saphenous veins. METHODS AND RESULTS Saphenous vein rings were obtained from 32 patients undergoing coronary artery bypass surgery. The vein rings were suspended in organ bath chambers for isometric recording of tension. AVP (3x10[-9] mol/L) enhanced the contractions elicited by electrical field stimulation at 1, 2, and 4 Hz (by 80%, 70%, and 60%, respectively) and produced a leftward shift of the concentration-response curve to norepinephrine (half-maximal effective concentration decreased from 6.87x10[-7] to 1.04x10[-7] mol/L; P<.05). The V1 vasopressin receptor antagonist d(CH2)5Tyr(Me)AVP (10[-6] mol/L) prevented the potentiation evoked by AVP. The selective V1 receptor agonist [Phe,2 Orn8]-vasotocin (3x[-10]-9 mol/L) induced potentiation of electrical stimulation-evoked responses, which was also inhibited in the presence of the V1 receptor antagonist (10[-6] mol/L). In contrast, the V2 receptor agonist desmopressin (10[-9] to 10[-7] mol/L) did not modify neurogenic responses, and the V2 receptor antagonist [d(CH2)5, D-Ile,2 Ile,4 Arg8]-vasopressin (10[-8] to 10[-6] mol/L) did not prevent the potentiation induced by AVP. The dihydropyridine calcium antagonist nifedipine (10[-6] mol/L) did not affect the potentiating effect of AVP. CONCLUSIONS The results suggest that low concentrations of AVP facilitate sympathetic neurotransmission and potentiate constrictor effects of norepinephrine in human saphenous veins. These effects appear to be mediated by V1 receptor stimulation and are independent of calcium entry through dihydropyridine calcium channels. Thus, AVP may contribute to vascular mechanisms involved in acute ischemic syndromes associated with venous grafts, particularly if the sympathetic nervous system is activated.


The Annals of Thoracic Surgery | 2000

Relaxation induced by cGMP phosphodiesterase inhibitors sildenafil and zaprinast in human vessels.

Pascual Medina; Gloria Segarra; Juan Martínez-León; José M. Vila; Martín Aldasoro; Eduardo Otero; Salvador Lluch

BACKGROUND Sildenafil is currently used in the treatment of erectile dysfunction. However, assessment of direct effects of sildenafil on coronary arteries and on arteries used as coronary grafts is unknown. This study was designed to investigate the effects of sildenafil on contracted human coronary, internal mammary, and radial arteries obtained from multiorgan donors. The observations were extended to forearm veins. Zaprinast was included in this study for comparison. METHODS Segments of left coronary, internal mammary, and radial arteries, and forearm veins were obtained from 16 multiorgan donors. Vascular rings were suspended in organ bath chambers and isometric tension was recorded. Then the effects of sildenafil, zaprinast, and sodium nitroprusside on precontracted vessels were studied. RESULTS Sildenafil (10(-8) - 3 x 10(-5) mol/L) caused concentration-dependent relaxation in the internal mammary arteries, radial arteries, and forearm veins. In the coronary arteries, sildenafil had a modest relaxant effect. In addition, sildenafil amplified the relaxation induced by sodium nitroprusside in all four vessels. Relaxation was unaffected by the inhibitor of nitric oxide synthase NG-monomethyl-L-arginine (10(-4) mol/L). Compared with zaprinast, sildenafil was eight to ten times more potent in terms of EC50 values. CONCLUSIONS The direct relaxant effects of sildenafil together with its synergistic interaction with nitric oxide donors should be considered in patients undergoing coronary bypass surgery, patients with low blood pressure, and patients receiving antihypertensive regimes.


European Journal of Pharmacology | 1993

Endothelium-dependent component in the contractile responses of human omental arteries to adrenergic stimulation

Martín Aldasoro; Carmen Martínez; J. M. Vila; Blas Flor; Salvador Lluch

The present study was designed to investigate the influence of endothelium-derived nitric oxide on the contractile responses of isolated human omental arteries to electrical field stimulation and noradrenaline. We measured isometric tension in artery rings obtained from portions of human omentum during the course of abdominal operations (32 patients). Electrical field stimulation induced frequency-dependent contractions which were abolished by tetrodotoxin (10(-6) M) and prazosin (10(-6) M), thus indicating that this effect was due to noradrenaline released from adrenergic nerves acting on alpha 1-adrenoceptors. The increases in tension induced by electrical field stimulation were of greater magnitude in arteries denuded of endothelium. NG-Nitro-L-arginine (L-NAME, 10(-4) M) potentiated the contractile response to electrical field stimulation in artery rings with endothelium but did not influence the contractile responses of endothelium-denuded arteries. The potentiation induced by L-NAME was completely reversed by L-arginine (10(-4) M), but not by D-arginine (10(-4) M). Contractile responses to noradrenaline were similar in arteries with and without endothelium. L-NAME (10(-4) M) had no significant effect on the contractile responses to noradrenaline. Our results suggest that electrical field stimulation releases endothelium-derived nitric oxide which inhibits the contractile responses of human omental arteries. The constrictor responses to noradrenaline are not modulated by the endothelium.


European Journal of Pharmacology | 1998

Vasopressin receptors involved in adrenergic neurotransmission in the circular muscle of the human vas deferens

Pascual Medina; Gloria Segarra; Pascual Chuan; Cristina Domenech; José M. Vila; Martín Aldasoro; Salvador Lluch

We studied the effects of vasopressin on the adrenergic responses of in vitro preparations of circular muscle from the vas deferens obtained from 28 men undergoing elective vasectomy. Vasopressin (3 x 10(-9)-3 x 10(-8) M) enhanced the phasic contractions elicited by electrical field stimulation and noradrenaline. This potentiation was blocked by the vasopressin V1 receptor antagonist d(CH2)5Tyr(Me)vasopressin (10(-6) M) but not by the vasopressin V2 receptor antagonist [d(CH2)5, D-Ile2,Ile4,Arg8]vasopressin (10(-6) M). The Ca2+ antagonist nifedipine (10(-6) M) did not affect the potentiation of electrical field stimulation induced by vasopressin and noradrenaline but reduced KCl-induced contractions and abolished the induction of phasic activity by vasopressin in the presence of KCl. The results demonstrate that vasopressin, in addition to its direct contractile effects, strongly potentiates contractions of human vas deferens elicited by adrenergic stimulation. Both the direct and indirect effects of vasopressin appear to be mediated by vasopressin V1 receptor stimulation and are independent of Ca2+ entry through dihydropyridine Ca2+ channels.


International Journal of Medical Sciences | 2015

Astrocytes protect neurons from Aβ1-42 peptide-induced neurotoxicity increasing TFAM and PGC-1 and decreasing PPAR-γ and SIRT-1.

Diana Aguirre-Rueda; Sol Guerra-Ojeda; Martín Aldasoro; Antonio Iradi; Elena Obrador; Angel Ortega; M. Dolores Mauricio; Jose Mª Vila; Soraya L. Valles

One of the earliest neuropathological events in Alzheimers disease is accumulation of astrocytes at sites of Aβ1-42 depositions. Our results indicate that Aβ1-42 toxic peptide increases lipid peroxidation, apoptosis and cell death in neurons but not in astrocytes in primary culture. Aβ1-42-induced deleterious neuronal effects are not present when neurons and astrocytes are mixed cultured. Stimulation of astrocytes with toxic Aβ1-42 peptide increased p-65 and decreased IκB resulting in inflammatory process. In astrocytes Aβ1-42 decreases protein expressions of sirtuin 1 (SIRT-1) and peroxisome proliferator-activated receptor γ (PPAR-γ) and over-expresses peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1) and mitochondrial transcription factor A (TFAM), protecting mitochondria against Aβ1-42-induced damage and promoting mitochondrial biogenesis. In summary our data suggest that astrocytes may have a key role in protecting neurons, increasing neural viability and mitochondrial biogenesis, acquiring better oxidative stress protection and perhaps modulating inflammatory processes against Aβ1-42 toxic peptide. This might be a sign of a complex epigenetic process in Alzheimers disease development.

Collaboration


Dive into the Martín Aldasoro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. M. Vila

University of Valencia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eva Serna

University of Valencia

View shared research outputs
Researchain Logo
Decentralizing Knowledge