Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Drugs R D ; 19(4): 319-328, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31452117

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension is a hemodynamic disorder. Signs and symptoms are generally difficult to recognize because they are non-specific. The current treatment for pulmonary arterial hypertension offers no cure or prevention; therefore, it is important to explore treatment avenues for novel pulmonary arterial hypertension treatments. In this study, we tested the hypothesis: pulmonary vasodilator responses of adenosine are dependent on the activation of L-type calcium channels, independent of the synthesis of nitric oxide from L-arginine, activation of adenosine triphosphate-sensitive potassium channels, and the release of cyclooxygenase products. METHODS: We performed an isolated lobar lung preparation in mongrel cats. The thromboxane A2 analog U-46619 was used to increase lobar arterial pressure to a high steady level. We recorded responses to adenosine and other vasodepressor agents in the pulmonary vascular bed of a cat under conditions of controlled pulmonary blood flow and constant left atrial pressure. RESULTS: These data show that adenosine has significant vasodepressor activity in the pulmonary vascular bed of the cat. The data suggest that pulmonary vasodilator responses to adenosine are partially dependent on the activation of adenosine 1 and 2 receptor pathways, and independent of the activation of cyclooxygenase activation, adenosine triphosphate-sensitive K + channels, or synthesis of nitric oxide in the pulmonary vascular bed of the cat. CONCLUSIONS: Vasodepressor effects of adenosine are species specific, and this species specificity will impact the development of future testing and treatments for pulmonary arterial hypertension. Clinical studies are warranted to see if adenosine moieties could play a therapeutic role in patients with pulmonary arterial hypertension and/or other pulmonary pathogeneses.


Subject(s)
Adenosine/therapeutic use , Calcium Channels, L-Type/metabolism , Hypertension, Pulmonary/drug therapy , Vasodilator Agents/therapeutic use , Animals , Cats , Disease Models, Animal , Female , Hypertension, Pulmonary/metabolism , Male , Nitric Oxide/metabolism , Potassium Channels/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Signal Transduction
2.
Am J Physiol Heart Circ Physiol ; 294(2): H688-98, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032519

ABSTRACT

The mechanism by which acetylcholine (ACh) decreases systemic arterial pressure and hindlimb vascular resistance was investigated in the anesthetized rat. ACh injections caused dose-dependent decreases in systemic arterial pressure and hindlimb vascular resistance. N(omega)-nitro-L-arginine methyl ester (L-NAME) had little effect on the magnitude of depressor and vasodilator responses but decreased response duration when baseline parameters were corrected by a nitric oxide (NO) donor infusion. The decrease in the duration of the ACh depressor response was prevented by the administration of excess L-arginine. The L-NAME-resistant component of the depressor response to ACh was attenuated by ebselen, a glutathione peroxidase mimic. The calcium-activated potassium (K(Ca)) antagonists charybdotoxin (ChTX) and apamin decreased the magnitude but not the duration of the hindlimb vasodilator response to ACh. The combination of L-NAME, ChTX, and apamin reduced the magnitude and duration of the vasodilator response to ACh but not to sodium nitroprusside. Vasodepressor and hindlimb vasodilator responses to ACh were not modified by cytochrome P-450 and cyclooxygenase pathway inhibitors. These results suggest that the hindlimb vasodilator response to ACh has an initial L-NAME-resistant component mediated by the activation of K(Ca) channels and a sustained L-NAME-dependent component. The results with ebselen suggest that the L-NAME-resistant component of the depressor response involves a peroxide-sensitive mechanism. The present study suggests that vasodilator responses to ACh are not mediated by cytochrome P-450 products, since miconazole and 1-aminobentriazole alone or in combination did not affect either component of the response. The present data suggest that the hindlimb vasodilator response to ACh in the rat is mediated by two mechanisms with an initial ChTX- and apamin-sensitive, L-NAME-resistant phase not mediated by cytochrome P-450 products and a secondary sustained phase mediated by NO.


Subject(s)
Acetylcholine/pharmacology , Enzyme Inhibitors/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Animals , Apamin/pharmacology , Blood Pressure/drug effects , Charybdotoxin/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Drug Resistance , Heart Rate/drug effects , Hindlimb/blood supply , Male , Muscle Tonus/drug effects , NG-Nitroarginine Methyl Ester/antagonists & inhibitors , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Peroxides/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/metabolism , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Vasodilator Agents/pharmacology
3.
J Cardiovasc Pharmacol ; 50(4): 358-66, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18049302

ABSTRACT

The free radical peroxynitrite (ONOO-) is formed in biological systems from the reaction of nitric oxide (NO) with superoxide (O2-) and can react with protein and nonprotein thiol groups to produce tissue injury. However, these pathologic actions of (ONOO-) may have been overemphasized, in that (ONOO-) has vasorelaxant properties through activation of soluble guanylate cyclase; inhibits leukocyte-endothelial cell interactions; and reduces ischemia-reperfusion injury in the heart, lung, and liver. It has been reported that tolerance develops to the vasodilator actions of (ONOO-) and that (ONOO-) impairs vascular function. However, little, if anything, is known about responses to (ONOO-) in the hindlimb circulation of the cat. To better understand the effects of (ONOO-) on responses to vasoactive agonists and the mechanism by which (ONOO-) induces vasodilation, the effects of short-term exposure to (ONOO-) were investigated under constant-flow conditions in the hindlimb vascular bed of the cat. In these studies, direct intraarterial injections of (ONOO-) produced dose-dependent decreases in hindquarters perfusion pressure. The vasodilator responses to (ONOO-) were rapid in onset, were short in duration, and could be repeated without exhibiting tachyphylaxis. Vasodilator responses to (ONOO-) were not changed in the presence of inhibitors of nitric-oxide synthase, cyclooxygenase, or K+-ATP (adenosine triphosphate-sensitive potassium) channels. Furthermore, responses to (ONOO-) were enhanced in duration by the type 5-cGMP (cyclic guanosine monophosphate) phosphodiesterase inhibitor zaprinast, whereas rolipram, a type 4-cGMP phosphodiesterase inhibitor, was without effect. Repeated administration of (ONOO-) had no significant effect on responses to vasoconstrictor or to vasodilator agents including acetylcholine. These results show that (ONOO-) has significant vasodilator activity in the hindlimb vascular bed of the cat and suggest that the response is mediated by a cGMP- dependent mechanism. The results of experiments with repeated injections of (ONOO-) indicate that (ONOO-) does not impair vasoconstrictor and endothelium-dependent or endothelium-independent vasodilator responses. Furthermore, tolerance did not develop with repeated short-term exposure to (ONOO-). Moreover, the results of experiments with inhibitors suggest that responses to (ONOO-) are not dependent on K-ATP (adenosine triphosphate-sensitive potassium) channel activation, increased NOS activity, or the formation of products in the cyclooxygenase pathway. The results of these studies are consistent with the hypothesis that (ONOO-) is rapidly converted in the hindlimb circulation to a substance that has the properties of an NO donor. These studies suggest that under physiologic conditions, the cytotoxic effects of (ONOO-) on a short-term basis may have been overemphasized.


Subject(s)
Hindlimb/blood supply , Nitric Oxide Donors/pharmacology , Peroxynitrous Acid/pharmacology , Regional Blood Flow/drug effects , Vascular Resistance/drug effects , Vasodilator Agents/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Cats , Female , Hindlimb/drug effects , Male , Meclofenamic Acid/pharmacology , Morpholines/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/pharmacology , Perfusion , Phosphodiesterase Inhibitors/pharmacology , Potassium Channel Blockers/pharmacology , Purinones/pharmacology , Rolipram/pharmacology , Sodium Nitrite/pharmacology , Splanchnic Circulation/drug effects , Vasoconstrictor Agents/pharmacology
4.
Pharmacology ; 80(4): 249-60, 2007.
Article in English | MEDLINE | ID: mdl-17627114

ABSTRACT

In contrast to nonsteroidal anti-inflammatory drugs (NSAIDs), the nonopioid analgesics phenacetin, acetaminophen and dipyrone exhibit weak anti-inflammatory properties. An explanation for this difference in pharmacologic activity was provided by the recent discovery of a new cyclooxygenase isoform, cyclooxygenase (COX)-3, that is reported to be inhibited by phenacetin, acetaminophen and dipyrone. However, COX-3 was found to be a spliced variant of COX-1 and renamed COX-1b. Although recent studies provide evidence for the existence of this new COX isoform, it is uncertain whether this COX-3 (COX-1b) isoform, or putative acetaminophen-sensitive pathway, plays a role in the generation of vasoactive prostaglandins. NSAIDs increase systemic blood pressure by inhibiting the formation of vasodilator prostanoids. Angiotensin II, norepinephrine and other vasoconstrictor agents have been reported to release prostaglandins. It is possible that this acetaminophen-sensitive pathway also modulates pressor responses to these vasoconstrictor agents. Therefore, the purpose of the present study was to determine whether this acetaminophen-sensitive pathway plays a role in the generation of vasoactive products of arachidonic acid or in the modulation of vasoconstrictor responses in the pulmonary and systemic vascular bed of the intact-chest rat. In the present study, the nonopioid analgesics did not attenuate changes in pulmonary or systemic arterial pressure in response to injections of the prostanoid precursor, arachidonic acid, to the thromboxane A(2) mimic, U46619, or to angiotensin II or norepinephrine. The results of the present study do not provide evidence in support of a role of a functional COX-3 (COX-1b) isoform, or an acetaminophen-sensitive pathway, in the generation of vasoactive prostanoids or in the modulation of responses to vasoconstrictor hormones in the intact-chest rat.


Subject(s)
Acetaminophen/pharmacology , Arachidonic Acid/pharmacology , Blood Pressure/drug effects , Dipyrone/pharmacology , Phenacetin/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Prostaglandin-Endoperoxide Synthases/physiology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology
5.
Can J Physiol Pharmacol ; 85(3-4): 360-71, 2007.
Article in English | MEDLINE | ID: mdl-17612645

ABSTRACT

Losartan has been reported to have inhibitory effects on thromboxane (TP) receptor-mediated responses. In the present study, the effects of 2 nonpeptide angiotensin II (AT1) receptor antagonists, losartan and candesartan, on responses to angiotensin II, the thromboxane A2 mimic, U46619, and norepinephrine were investigated and compared in the pulmonary and systemic vascular beds of the intact-chest rat. In this study, intravenous injections of angiotensin II, U46619, and norepinephrine produced dose-related increases in pulmonary and systemic arterial pressure. Losartan and candesartan, in the doses studied, decreased or abolished responses to angiotensin II. Losartan, but not candesartan, and only in a higher dose, produced small, but statistically significant, reductions in pressor responses to U46619 and to norepinephrine in the pulmonary and systemic vascular beds. Furthermore, losartan significantly reduced arachidonic acid-induced platelet aggregation, whereas candesartan had no effect. Pressor responses to angiotensin II were not changed by thromboxane and alpha-adrenergic receptor antagonists, or by cyclooxygenase and NO synthase inhibitors. These results show that losartan and candesartan are potent selective AT1 receptor antagonists in the pulmonary and systemic vascular beds and that losartan can attenuate thromboxane and alpha-adrenergic responses when administered at a high dose, whereas candesartan in the highest dose studied had no effect on responses to U46619 or to norepinephrine. The present data show that the effects of losartan and candesartan on vasoconstrictor responses are different and that pulmonary and systemic pressor responses to angiotensin II are not modulated or mediated by the release of cyclooxygenase products, activation of TP receptors, or the release of NO in the anesthetized rat.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Losartan/pharmacology , Pulmonary Artery/drug effects , Tetrazoles/pharmacology , Vasoconstriction/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adrenergic alpha-Agonists/pharmacology , Angiotensin II/pharmacology , Animals , Biphenyl Compounds , Blood Pressure/drug effects , Cyclooxygenase Inhibitors/pharmacology , Meclofenamic Acid/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Norepinephrine/pharmacology , Platelet Aggregation/drug effects , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology
6.
Am J Physiol Heart Circ Physiol ; 292(2): H1120-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16980338

ABSTRACT

The administration of mesenchymal stem cells (MSCs) has been proposed for the treatment of pulmonary hypertension. However, the effect of intratracheally administered MSCs on the pulmonary vascular bed in monocrotaline-treated rats has not been determined. In the present study, the effect of intratracheal administration of rat MSCs (rMSCs) on monocrotaline-induced pulmonary hypertension and impaired endothelium-dependent responses were investigated in the rat. Intravenous injection of monocrotaline increased pulmonary arterial pressure and vascular resistance and decreased pulmonary vascular responses to acetylcholine without altering responses to sodium nitroprusside and without altering systemic responses to the vasodilator agents when responses were evaluated at 5 wk. The intratracheal injection of 3 x 10(6) rMSCs 2 wk after administration of monocrotaline attenuated the rise in pulmonary arterial pressure and pulmonary vascular resistance and restored pulmonary responses to acetylcholine toward values measured in control rats. Treatment with rMSCs decreased the right ventricular hypertrophy induced by monocrotaline. Immunohistochemical studies showed widespread distribution of lacZ-labeled rMSCs in lung parenchyma surrounding airways in monocrotaline-treated rats. Immunofluorescence studies revealed that transplanted rMSCs retained expression of von Willebrand factor and smooth muscle actin markers specific for endothelial and smooth muscle phenotypes. However, immunolabeled cells were not detected in the wall of pulmonary vessels. These data suggest that the decrease in pulmonary vascular resistance and improvement in response to acetylcholine an endothelium-dependent vasodilator in monocrotaline-treated rats may result from a paracrine effect of the transplanted rMSCs in lung parenchyma, which improves vascular endothelial function in the monocrotaline-injured lung.


Subject(s)
Endothelium, Vascular/physiopathology , Hypertension, Pulmonary/surgery , Mesenchymal Stem Cell Transplantation , Pulmonary Circulation , Vasodilation , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Lung/blood supply , Lung/pathology , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Monocrotaline , Paracrine Communication , Phenotype , Pulmonary Circulation/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Trachea/surgery , Vascular Resistance , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
J Cardiothorac Vasc Anesth ; 20(5): 691-5, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17023290

ABSTRACT

OBJECTIVE: The purpose of this study was to test the hypothesis that meperidine induces a dilator response in the feline pulmonary vascular bed, and to identify receptors involved in the mediation or modulation of these effects. DESIGN: Prospective vehicle controlled study. SETTING: University research laboratory. SUBJECTS: Intact chest preparation; adult mongrel cats. INTERVENTIONS: In separate experiments, the effects of diphenydramine (histamine H(1)-receptor antagonist), glibenclamide (adenosine triphosphate-sensitive K+ channel blocker), L-N(5)-(1-Iminoethyl) ornithine hydrochloride (L-NIO) (nitric oxide synthase inhibitor), naloxone (opioid receptor antagonist), and nimesulide (selective cyclooxygenase-2 inhibitor) were investigated on pulmonary arterial responses to meperidine and other agonists in the feline lung bed. MEASUREMENTS AND MAIN RESULTS: The systemic pressure and lobar arterial perfusion pressure were continuously monitored, electronically averaged, and permanently recorded. Under elevated tone conditions in the isolated left lower lobe vascular bed of the cat, meperidine induced a dose-dependent vasodilator response that was not significantly altered after administration of glibenclamide, L-NIO, and nimesulide. Responses to meperidine were significantly attenuated after the administration of diphenydramine and naloxone. CONCLUSIONS: The results suggest that meperidine has potent vasodilator activity in the feline pulmonary vascular bed, and these responses are mediated or modulated, in part, by opioid and histamine receptor-sensitive pathways.


Subject(s)
Analgesics, Opioid/pharmacology , Meperidine/pharmacology , Pulmonary Artery/drug effects , Pulmonary Veins/drug effects , Pulmonary Wedge Pressure/physiology , Vasodilation/physiology , Animals , Cats , Female , Follow-Up Studies , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypertension, Pulmonary/veterinary , Male , Prognosis , Prospective Studies , Pulmonary Artery/physiology , Pulmonary Veins/physiology , Pulmonary Wedge Pressure/drug effects , Vasodilation/drug effects
8.
Eur J Pharmacol ; 534(1-3): 159-64, 2006 Mar 18.
Article in English | MEDLINE | ID: mdl-16507307

ABSTRACT

The purpose of this prospective vehicle controlled study was to test the hypothesis that sufentanil induces a depressor response in the pulmonary vascular bed of the cat and identify the receptors involved in the mediation or modulation of these effects. In separate experiments, the effects of diphenydramine (histamine receptor blocker), glibenclamide (ATP-sensitive K+ channel blocker), L-N5-(1-Iminoethyl) ornithine hydrochloride (L-NIO) (nitric oxide synthase inhibitor), nimesulide (selective cyclooxygenase (COX)-2 inhibitor), and naloxone (opiate receptor antagonist) were investigated on pulmonary arterial responses to sufentanil and other agonists in the feline pulmonary vascular bed. The lobar arterial perfusion pressures were continuously monitored, electronically averaged, and recorded. In the feline pulmonary vascular bed of the isolated left lower lobe, sufentanil induced a dose-dependent vasodepressor response that was not significantly altered after administration of glibenclamide, L-NIO, and nimesulide. However, the responses to sufentanil were significantly attenuated following administration of diphenhydramine and naloxone. The results of the present study suggest that sufentanil has potent vasodepressor activity in the pulmonary vascular bed of the cat and that this response may be mediated or modulated by both histaminergic and opioid receptor sensitive pathways.


Subject(s)
Analgesics, Opioid/pharmacology , Pulmonary Artery/drug effects , Sufentanil/pharmacology , Vasodilation , Vasodilator Agents/pharmacology , Animals , Cats , Diphenhydramine/pharmacology , Dose-Response Relationship, Drug , Histamine Antagonists/pharmacology , Lung/blood supply , Lung/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pulmonary Circulation , Vascular Resistance
9.
Am J Ther ; 13(1): 12-7, 2006.
Article in English | MEDLINE | ID: mdl-16428917

ABSTRACT

In a university research laboratory and in separate experiments, the effects of phentolamine, the alpha-adrenergic antagonist; prazosin, an alpha1-adrenoceptor antagonist; 5-methyl-urapidil, the selective alpha1A-subtype adrenoceptor antagonist; chloroethylclonidine, an alpha1B- and alpha1D-subtype adrenoceptor antagonist; and BMY 7378, a selective alpha1D-subtype adrenoceptor antagonist were analyzed in an attempt to identify any significant effect on pulmonary arterial responses to ephedrine and other agonist agents in the pulmonary vascular bed of the cat. Under constant flow conditions, lobar arterial perfusion pressure and systemic pressure were continuously monitored, electronically averaged, and permanently recorded. In the isolated left lower lobe of the pulmonary feline vascular bed, ephedrine induced a dose-dependent vasoconstrictor response that was not significantly altered following administration of 5-methyl-urapidil. The vasopressor activity as a result of ephedrine was significantly decreased after administration of phentolamine, prazosin, chloroethylclonidine, and BMY 7378. Further, when the alpha1B- and alpha1D-subtype adrenoceptor antagonist chloroethylclonidine was given, there was almost complete elimination of the ephedrine-induced vasoconstrictor response. The results of this study suggest that ephedrine causes a dose-dependent vasopressor response in the feline pulmonary vascular bed and that this activity may be mediated or modulated by both alpha1B- and alpha1D-subtype adrenoceptor sensitive pathways.


Subject(s)
Ephedrine/pharmacology , Lung/blood supply , Receptors, Adrenergic, alpha-1/physiology , Adrenergic alpha-1 Receptor Agonists , Adrenergic alpha-1 Receptor Antagonists , Animals , Cats , Dose-Response Relationship, Drug , Female , Male , Microcirculation/drug effects , Vasoconstriction/drug effects
10.
J Pharmacol Exp Ther ; 316(1): 95-105, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16002460

ABSTRACT

The mechanism of the pressor response to ephedrine is controversial. In the present study. i.v. injections of ephedrine increased systemic and pulmonary arterial pressure, and i.a. injections decreased hindlimb blood flow in a dose-related manner. Responses to ephedrine were inhibited by alpha-receptor blocking agents and were not attenuated by blockade of the norepinephrine reuptake transporter (NET) or by catecholamine depletion using reserpine or a combination of reserpine and alpha-methyl-p-tyrosine, whereas responses to tyramine and amphetamine were inhibited by these treatments. The magnitude of the pressor response to ephedrine was similar in anesthetized and conscious rats. Tachyphylaxis developed to pressor responses to ephedrine and amphetamine with sequential injections; however, ephedrine tachyphylaxis differed in that subsequent responses to alpha-receptor agonists were attenuated. These results suggest that the systemic and pulmonary pressor and hindlimb vasoconstrictor responses to ephedrine are mediated by direct action on alpha-adrenergic receptors and that the release of norepinephrine from adrenergic terminals plays no significant role. These results provide support for the hypothesis that responses to ephedrine are directly mediated in the intact rat, whereas responses to amphetamine are mediated in a large part by the release of norepinephrine from adrenergic terminals.


Subject(s)
Blood Pressure/drug effects , Central Nervous System Stimulants/pharmacology , Ephedrine/pharmacology , Amphetamine/pharmacology , Anesthesia , Animals , Catecholamines/metabolism , Chromatography, High Pressure Liquid , Cocaine/pharmacology , Hindlimb/blood supply , Male , Norepinephrine/pharmacology , Phenylephrine/pharmacology , Pulmonary Artery/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/drug effects , Regional Blood Flow/drug effects , Reserpine/pharmacology
11.
Anesth Analg ; 102(1): 118-23, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16368815

ABSTRACT

In this investigation we sought to identify the role of remifentanil in the feline pulmonary vascular bed. Using adult mongrel cats in separate experiments, the effects of glibenclamide (adenosine triphosphate-sensitive K+ channel blocker), diphenhydramine (histamine H(1)-receptor antagonist), L-N5-(1-Iminoethyl) ornithine hydrochloride (nitric oxide synthase inhibitor), and naloxone (opioid receptor antagonist) were investigated in pulmonary arterial responses to remifentanil (opioid agonist), pinacidil (adenosine triphosphate-sensitive K+ channel activator), and bradykinin (nitric oxide synthase inducer). Under increased tone conditions in the isolated left lower lobe vascular bed of the cat, remifentanil induced a dose-dependent vasodepressor response that was not significantly altered after administration of glibenclamide and L-N5-(1-Iminoethyl) ornithine hydrochloride. Responses to remifentanil were significantly attenuated after administration of diphenhydramine and naloxone. The results suggest that remifentanil has potent vasodepressor activity in the feline pulmonary vascular bed and that these responses are mediated by histamine and opioid receptor sensitive pathways.


Subject(s)
Lung/blood supply , Lung/drug effects , Piperidines/pharmacology , Pulmonary Circulation/drug effects , Animals , Cats , Dose-Response Relationship, Drug , Female , Lung/physiology , Male , Pulmonary Circulation/physiology , Remifentanil , Vascular Resistance/drug effects , Vascular Resistance/physiology , Vasodilation/drug effects , Vasodilation/physiology
12.
Can J Physiol Pharmacol ; 84(11): 1163-75, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17218981

ABSTRACT

Angiotensin-(1-7) (Ang-(1-7)), a bioactive peptide in the renin-angiotensin system, has counterregulatory actions to angiotensin II (Ang II). However, the mechanism by which Ang-(1-7) enhances vasodepressor responses to bradykinin (BK) is not well understood. In the present study, the effects of Ang-(1-7) on responses to BK, BK analogs, angiotensin I (Ang I), and Ang II were investigated in the anesthetized rat. The infusion of Ang-(1-7) (55 pmol/min i.v.) enhanced decreases in systemic arterial pressure in response to i.v. injections of BK and the BK analogs [Hyp3, Tyr(Me)8]-bradykinin (HT-BK) and [Phe8psi (CH2-NH) Arg9]-bradykinin (PA-BK) without altering pressor responses to Ang I or II, or depressor responses to acetylcholine and sodium nitroprusside. The angiotensin-converting enzyme (ACE) inhibitor enalaprilat enhanced responses to BK and the BK analog HT-BK without altering responses to PA-BK and inhibited responses to Ang I. The potentiating effects of Ang-(1-7) and enalaprilat on responses to BK were not attenuated by the Ang-(1-7) receptor antagonist A-779. Ang-(1-7)- and ACE inhibitor-potentiated responses to BK were attenuated by the BK B2 receptor antagonist Hoe 140. The cyclooxygenase inhibitor sodium meclofenamate had no significant effect on responses to BK or Ang-(1-7)-potentiated BK responses. These results suggest that Ang-(1-7) potentiates responses to BK by a selective B2 receptor mechanism that is independent of an effect on Ang-(1-7) receptors, ACE, or cyclooxygenase product formation. These data suggest that ACE inhibitor-potentiated responses to BK are not mediated by an A-779-sensitive mechanism and are consistent with the hypothesis that enalaprilat-induced BK potentiation is due to decreased BK inactivation.


Subject(s)
Angiotensin I/pharmacology , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Bradykinin/pharmacology , Peptide Fragments/pharmacology , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bradykinin/analogs & derivatives , Carotid Artery, External/drug effects , Cyclooxygenase Inhibitors , Dose-Response Relationship, Drug , Drug Synergism , Enalaprilat/pharmacology , Male , Meclofenamic Acid/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B2/drug effects , Receptors, G-Protein-Coupled/drug effects
13.
Can J Physiol Pharmacol ; 83(7): 529-39, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16091779

ABSTRACT

Stem cells isolated from various sources have been shown to vary in their differentiation capacity or pluripotentiality. Two groups of stem cells, embryonic and adult stem cells, may be capable of differentiating into any desired tissue or cell type, which offers hope for the development of therapeutic applications for a large number of disorders. However, major limitations with the use of embryonic stem cells for human disease have led researchers to focus on adult stem cells as therapeutic agents. Investigators have begun to examine postnatal sources of pluripotent stem cells, such as bone marrow stroma or adipose tissue, as sources of mesenchymal stem cells. The following review focuses on recent research on the use of stem cells for the treatment of cardiovascular and pulmonary diseases and the future application of mesenchymal stem cells for the treatment of a variety of cardiovascular disorders.


Subject(s)
Cardiovascular Diseases/therapy , Mesoderm/cytology , Stem Cell Transplantation , Stem Cells/physiology , Animals , Erectile Dysfunction/therapy , Humans , Lung Diseases/therapy , Male
14.
J Appl Toxicol ; 25(6): 479-90, 2005.
Article in English | MEDLINE | ID: mdl-16092079

ABSTRACT

Chronic cocaine abuse is known to cause endothelial dysfunction and atherosclerosis. The present study investigated the effect of binge cocaine treatment, a model for chronic cocaine abuse, on the blood flow responses to the adrenergic agonists norepinephrine, phenylephrine and isoproterenol, the endothelium-dependent vasodilator acetylcholine, and the endothelium independent vasodilator sodium nitroprusside (SNP) in the hindlimb vascular bed of male Sprague Dawley rats. Rats received either single binge or double binge treatment. Each binge treatment consisted of three doses of cocaine (30 mg kg(-1) i.p.) for 3 days. For double binge treatment, there was a 4 day recovery period between the binges. At the end of the treatment the rats were anesthetized and agonists were administered into the right hindlimb circulation through a catheter in the left iliac artery and blood flow responses were measured with a flow probe around the right iliac artery. Rats receiving double cocaine binges showed a significant decrease in the magnitude and duration of the blood flow response to norepinephrine and a decrease in the duration of the blood flow response to phenylephrine, isoproterenol and acetylcholine when compared with saline controls. The blood flow response to SNP was not changed. Total plasma nitrate-nitrite levels were significantly reduced and big endothelin levels were significantly increased in rats receiving double cocaine binges. This study demonstrates that binge cocaine treatment can alter endothelial function, while not changing smooth muscle function, and impairs the adrenergic pathway.


Subject(s)
Cocaine-Related Disorders/physiopathology , Cocaine/toxicity , Hindlimb/blood supply , Acetylcholine , Animals , Bradykinin , Cocaine/administration & dosage , Cocaine-Related Disorders/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelin-1/blood , Endothelium, Vascular/drug effects , Hindlimb/physiopathology , Male , Nitroprusside , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Time Factors , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
15.
Am J Physiol Heart Circ Physiol ; 289(4): H1476-87, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15863457

ABSTRACT

Generation of vasoactive prostanoids from arachidonic acid by cyclooxygenase (COX)-1 and COX-2 was investigated in anesthetized mice. Intravenous injections of the prostanoid precursor arachidonic acid increased pulmonary arterial pressure and decreased systemic arterial pressure. Pulmonary pressor and systemic depressor responses were attenuated by SC-560 and nimesulide, inhibitors of COX-1 and COX-2, in doses that did not alter responses to injected prostanoids. Pulmonary pressor responses to arachidonic acid were blocked and a depressor response was unmasked, whereas systemic depressor responses were not altered, by a thromboxane receptor antagonist. Pulmonary and systemic pressor responses to angiotensin II injections and systemic pressor responses to angiotensin II infusion were not modified by COX-1 or COX-2 inhibitors but were attenuated by losartan. Systemic depressor responses to arachidonic acid were smaller in COX-1 and COX-2 knockout mice, whereas responses to angiotensin II, norepinephrine, U-46619, endothelin-1, and PGE(1) were not different in COX-1 and COX-2 knockout and wild-type control mice. These results suggest that vasoactive prostanoids with pulmonary pressor and systemic vasodepressor activity are formed by COX-1 and COX-2 and are consistent with Western blot analysis and immunostaining showing the presence of COX-1 and COX-2. These data suggest that thromboxane A(2) (TxA(2)) is formed from the precursor by COX-1 and COX-2 in the lung and are in agreement with immunofluorescence studies showing thromboxane synthase. The present data suggest that COX-1- or COX-2-derived prostanoids do not modulate responses to angiotensin II or other vasoactive agents and that prostanoid responses are similar in CD-1 and C57BL/6 and in male and female mice.


Subject(s)
Arachidonic Acid/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Pulmonary Circulation/physiology , Thromboxane A2/metabolism , Angiotensin II/pharmacology , Animals , Arachidonic Acid/pharmacology , Cyclooxygenase 1 , Cyclooxygenase 2 , Female , Gene Expression Regulation, Enzymologic , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/genetics , Pulmonary Circulation/drug effects , Thromboxane A2/biosynthesis , Thromboxane-A Synthase/genetics , Thromboxane-A Synthase/metabolism , Vasoconstrictor Agents/pharmacology
16.
J Cardiovasc Pharmacol ; 45(5): 431-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15821438

ABSTRACT

Chronic alveolar hypoxia induces vascular changes leading to pulmonary hypertension. We investigated the role of nitric oxide synthase (NOS) on basal pulmonary artery pressure (PAP) and on changes in PAP arising from an acute alveolar hypoxic challenge (AAHC) in normoxic and chronically hypoxic young rabbits. The chronically hypoxic rabbits were raised from birth in a chamber containing a (10% O2 + 90% N2) gas mixture, whereas the normoxic rabbits were kept in room air. The age of the animals at the time of study (approximately 38 days) was not significantly different between the 2 groups of rabbits. The in vivo PAP was measured using a right heart catheterization technique while the rabbits were spontaneously breathing either a hyperoxic or a hypoxic gas. In the chronically hypoxic group, the AAHC (hypoxic gas) produced a modest increase in PAP. However, after intravenous administration of (100 mg/kg) of the NOS inhibitor, L-NAME (N-nitro-L-arginine methyl ester), a marked increase in PAP was observed when these rabbits were rechallenged with the AAHC. In contrast, in the normoxic rabbits, the AAHC produced only a small increase in PAP, even after pretreatment with L-NAME. In both groups of rabbits, L-NAME led to a significant rise in basal PAP. Using Western blot analysis, we found endothelial NOS (eNOS) protein expression to be significantly increased in pulmonary artery and right ventricular myocardium of the chronically hypoxic rabbits. These results suggest that release of nitric oxide is involved in regulating basal PAP and in modulating the hypoxia-induced pulmonary vasoconstrictor response in immature animals. Moreover, eNOS appears to undergo up-regulation as a consequence of chronic hypoxia.


Subject(s)
Hypoxia/physiopathology , Lung/blood supply , Lung/physiopathology , Animals , Blood Pressure/drug effects , Blotting, Western , Enzyme Inhibitors/pharmacology , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Hypertension, Pulmonary/physiopathology , Lung/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Rabbits , Time Factors
17.
Anesth Analg ; 99(3): 758-63, table of contents, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15333407

ABSTRACT

In this investigation, we sought to identify the role of gamma-aminobutyric acid (GABA)(A) and GABA(B) receptors in the feline pulmonary vascular bed. Using adult mongrel cats and in separate experiments, we investigated the effects of l-N(5)-(1-iminoethyl) ornithine hydrochloride (l-NIO) (a nitric oxide synthase inhibitor), glibenclamide (an adenosine triphosphate (ATP)-sensitive K(+) channel blocker), meclofenamate (a nonselective cyclooxygenase inhibitor), bicuculline (a GABA(A) receptor antagonist), and saclofen (a GABA(B) receptor antagonist) on pulmonary arterial responses to pinacidil (an ATP-sensitive K(+) channel activator), bradykinin (a nitric oxide synthase inducer), muscimol (a GABA(A) receptor agonist), and 3-aminopropyl(methyl)phosphinic acid, hydrochloride (SKF-97541; a GABA(B) receptor agonist). Under increased tone conditions in the isolated left lower lobe vascular bed of the cat, muscimol induced a dose-dependent vasodepressor response that was not significantly altered after the administration of l-NIO, glibenclamide, meclofenamate, and saclofen. SKF-97541-induced vasodepression was not significantly attenuated after the administration of l-NIO, meclofenamate, and bicuculline. Responses to SKF-97541 were significantly attenuated after the administration of glibenclamide and saclofen. Responses to muscimol were significantly reduced after the administration of bicuculline. The results suggest that muscimol and SKF-97541 have potent vasodepressor activity in the feline pulmonary vascular bed and that these responses are modulated by, respectively, GABA(A) and GABA(B) receptor-sensitive pathways. Further, SKF-97541-induced vasodepression is mediated or modulated by an ATP-sensitive K(+) channel.


Subject(s)
Baclofen/analogs & derivatives , Lung/drug effects , Receptors, GABA-A/physiology , Receptors, GABA-B/physiology , Animals , Baclofen/pharmacology , Blood Vessels/drug effects , Blood Vessels/physiology , Cats , Dose-Response Relationship, Drug , Female , Glyburide/pharmacology , Lung/blood supply , Male , Meclofenamic Acid/pharmacology , Organophosphorus Compounds/pharmacology
18.
Anesth Analg ; 99(2): 758-763, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15271700

ABSTRACT

Tissue Doppler Imaging (TDI) provides information on systolic function through its systolic mitral annulus velocity wave (Sm), reflecting the peak velocity of shortening of the myocardial fibers oriented in the longitudinal direction. In this study, we evaluated the effect of load changes on Sm. Forty-two cardiac surgical patients with left ventricular ejection fraction >60% were consecutively evaluated. In 24 patients, load was changed with an IV bolus of phenylephrine (50-100 microg) or nitroglycerine (300-500 microg); in 18 patients, preload was changed with a rapid infusion of 500 mL of a gelatin solution. The sample volume of TDI was placed at the lateral side of the mitral annulus in the mid-esophageal 4-chamber view. Changing loading conditions with phenylephrine or nitroglycerine had no effect on Sm; the increase of preload in 18 patients resulted in a statistically significant increase of Sm (baseline, 8.4 +/- 2.6 cm/s; after increase of preload, 9.6 +/- 2.5 cm/s; P = 0.001). We conclude that Sm is dependent on changes in preload obtained by volume loading and cannot be recommended as an index of ventricular contractile performance in critically ill patients where significant changes in ventricular filling occur.


Subject(s)
Echocardiography, Transesophageal , Mitral Valve/diagnostic imaging , Mitral Valve/physiology , Myocardial Contraction/physiology , Aged , Coronary Artery Bypass , Electrocardiography , Female , Heart Rate/drug effects , Heart Rate/physiology , Hemodynamics/physiology , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Nitroglycerin/pharmacology , Phenylephrine/pharmacology , Systole/physiology , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Ventricular Function , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
19.
Crit Care Med ; 32(11): 2300-3, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15640645

ABSTRACT

OBJECTIVE: To test the hypothesis that norepinephrine induces a pressor response in the pulmonary vascular bed of the cat and identify the alpha-(1)adrenoceptor subtypes involved in the mediation or modulation of these effects. DESIGN: Prospective vehicle controlled study. SETTING: University research laboratory. SUBJECTS: Intact chest preparation, adult mongrel cats. INTERVENTIONS: In separate experiments, the effects of 5-methyl-urapidil, a selective alpha-(1)A-subtype adrenoceptor antagonist, chloroethylclonidine, an alpha-(1)B-subtype and -(1)D-subtype adrenoceptor antagonist, and BMY 7378, the selective alpha-(1)D-subtype adrenoceptor antagonist, were investigated on pulmonary arterial responses to norepinephrine and other agonists in the pulmonary vascular bed of the cat. MEASUREMENTS AND MAIN RESULTS: The systemic pressure and lobar arterial perfusion pressure were continuously monitored, electronically averaged, and permanently recorded. In the feline pulmonary vascular bed of the isolated left lower lobe, norepinephrine induced a dose-dependent vasoconstrictor response that was not significantly altered after administration of BMY 7378. However, the responses to norepinephrine were significantly attenuated following administration of 5-methyl-urapidil and chloroethylclonidine. CONCLUSIONS: The results of the present study suggest that norepinephrine has potent vasopressor activity in the pulmonary vascular bed of the cat and that this response may be mediated or modulated by both alpha-(1)A-subtype and -(1)B-subtype adrenoceptor sensitive pathways.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Clonidine/analogs & derivatives , Norepinephrine/pharmacology , Pulmonary Artery/drug effects , Pulmonary Circulation/drug effects , Receptors, Adrenergic, alpha-1/drug effects , Vasoconstrictor Agents/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Angiotensin II/pharmacology , Animals , Cats , Clonidine/pharmacology , Dose-Response Relationship, Drug , Drug Antagonism , Drug Evaluation, Preclinical , Ephedrine/pharmacology , Female , Injections, Intra-Arterial , Injections, Intravenous , Male , Phenylephrine/pharmacology , Piperazines/pharmacology , Receptors, Adrenergic, alpha-1/physiology , Vascular Resistance/drug effects
20.
Am J Physiol Heart Circ Physiol ; 285(6): H2399-410, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14613912

ABSTRACT

The role of cyclooxygenase (COX)-1 and -2 in prostanoid formation and modulation of pressor responses to ANG II was investigated in the pulmonary and systemic vascular beds in the rat. In the present study, selective COX-1 and -2 inhibitors attenuated increases in pulmonary arterial pressure and decreases in systemic arterial pressure in response to arachidonic acid but did not alter responses to PGE1 or U-46619. The selective COX-1 and -2 inhibitors did not modify systemic pressor responses to injections or infusions of ANG II or pulmonary pressor responses to injections of the peptide. COX-2 inhibitors did not alter, whereas a COX-1 inhibitor depressed, arachidonic acid-induced platelet aggregation. These data provide evidence in support of the hypothesis that prostanoid synthesis occurs by way of the COX-1 and -2 pathways in the pulmonary and systemic vascular beds but that pressor responses to ANG II are not mediated or modulated by these pathways in the rat.


Subject(s)
Angiotensin II/pharmacology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis , Pulmonary Circulation/physiology , Vasoconstrictor Agents/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Alprostadil/pharmacology , Animals , Arachidonic Acid/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiac Catheterization/methods , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Membrane Proteins , Nitrobenzenes/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Pulmonary Circulation/drug effects , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...